EP2035827B1 - Acide nucléique, polypeptide et son utilisation - Google Patents

Acide nucléique, polypeptide et son utilisation Download PDF

Info

Publication number
EP2035827B1
EP2035827B1 EP07720194A EP07720194A EP2035827B1 EP 2035827 B1 EP2035827 B1 EP 2035827B1 EP 07720194 A EP07720194 A EP 07720194A EP 07720194 A EP07720194 A EP 07720194A EP 2035827 B1 EP2035827 B1 EP 2035827B1
Authority
EP
European Patent Office
Prior art keywords
csr
cells
protein
seq
chimeric protein
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Active
Application number
EP07720194A
Other languages
German (de)
English (en)
Other versions
EP2035827A1 (fr
Inventor
Jay Patrick Slack
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Givaudan SA
Original Assignee
Givaudan SA
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Givaudan SA filed Critical Givaudan SA
Publication of EP2035827A1 publication Critical patent/EP2035827A1/fr
Application granted granted Critical
Publication of EP2035827B1 publication Critical patent/EP2035827B1/fr
Active legal-status Critical Current
Anticipated expiration legal-status Critical

Links

Classifications

    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/5005Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving human or animal cells
    • G01N33/5008Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving human or animal cells for testing or evaluating the effect of chemical or biological compounds, e.g. drugs, cosmetics
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/705Receptors; Cell surface antigens; Cell surface determinants
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/68Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving proteins, peptides or amino acids
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2319/00Fusion polypeptide
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2319/00Fusion polypeptide
    • C07K2319/32Fusion polypeptide fusions with soluble part of a cell surface receptor, "decoy receptors"
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N2333/00Assays involving biological materials from specific organisms or of a specific nature
    • G01N2333/435Assays involving biological materials from specific organisms or of a specific nature from animals; from humans
    • G01N2333/705Assays involving receptors, cell surface antigens or cell surface determinants
    • G01N2333/72Assays involving receptors, cell surface antigens or cell surface determinants for hormones
    • G01N2333/726G protein coupled receptor, e.g. TSHR-thyrotropin-receptor, LH/hCG receptor, FSH

Definitions

  • Sweetness modulators and in particular sweetness enhancers are of great interest to the food and flavor industry, for example, to allow reduction of sweeteners including sugars or artificial sweeteners.
  • the use of sweetness enhancers can reduce calories, prevent teeth from damage by sugars, and avoid or reduce the bitter/metallic off- and aftertastes associated with many artificial sweeteners.
  • T1 R2/T1 R3 heterodimeric sweet receptor To screen for sweetness modifiers or enhancers, known screens employing the T1 R2/T1 R3 heterodimeric sweet receptor can be used.
  • WO 03/004992 discloses T1 R2 and/or T1 R3 polypeptides and their use in methods to screen for sweetness modifiers or enhancers.
  • a sweetness modifier/enhancer To identify or characterise a sweetness modifier/enhancer, usually the results of samples with and without potential enhancer/modifier, both samples additionally containing a sweetener, are compared.
  • sweeteners and in particular sugars have a great effect on osmolarity, and/or are viscous. Due to changes in properties of the samples such as viscosity and osmolarity, artifacts may occur that cause incorrect results when using standard screening methods.
  • T1 R2/T1 R3 receptor comprises several binding domains, in particular the extracellular amino terminal domains including the venus flytrap ("VFT") domain that bind to carbohydrate sweeteners such as sucrose, glucose, fructose as well as the artificial sweeteners aspartame and sucralose. Therefore, a screen for specific modulators of specific ligands, and in particular for ligands of the transmembrane domains ("TMD(s)”) and cystein-rich domains of T1 R2 and/or T1 R3, excluding the VFT ligands, is not possible with known screening methods.
  • VFT venus flytrap
  • Agonists that bind in the TMD of T1 R3 are cyclamate and Neohesperidin Dihydrochalcone (NDHC). Sucrose and sucralose bind in the VFTs of T1 R2 and T1 R3, asparatame binds in the VFT of T1 R2.
  • NDHC Neohesperidin Dihydrochalcone
  • a CSR::T1R chimeric protein able to bind to at least one sweetener or sweetness enhancer, comprising one or more CSR::T1R selected from the group consisting of a CSR::T1R2 polypeptide substantially homologous to SEQ ID NO:2 (CSR::T1R2-a) or SEQ ID NO:20 (CSR::T1R2-b)with a sequence identity of at least 90%, a CSR:: T1R3 polypeptide substantially homologous to SEQ ID NO:4 (CSR::T1R3-a) or SEQ ID NO: 22 (CSR::T1R3-b) with a sequence identity of at least 90%.
  • a CSR::T1R chimeric protein as defined herein-above comprising two polypeptide subunits in form of a heterodimeric protein selected from the group consisting of a CSR::T1 R2/CSR::T1 R3 heterodimeric chimeric protein, a CSR::T1R2/T1R3 heterodimeric chimeric protein, and a T1R2/CSR::T1R3 heterodimeric chimeric protein, wherein the T1 R2 subunit of the heterodimer comprises a polypeptide a polypeptide essentially homologous to SEQ ID NO:8 with a sequence identity of at least 90%; and wherein the T1 R3 subunit of the heterodimer comprises a polypeptide essentially homologous to SEQ ID NO:10 with a sequence identity of at least 90%.
  • a CSR::T1R chimeric protein comprising two polypeptide subunits as defined herein-above which is a CSR::T1R2/CSR::T1 R3 heterodimeric chimeric protein including but not limited to a CSR::T1R2-a/CSR::T1R3-a heterodimeric protein, a CSR::T1 R2-b/CSR::T1 R3-b heterodimeric protein, a CSR::T1 R2-a/CSR::T1 R3-b heterodimeric protein, a CSR::T1 R2-b/CSR::T1 R3-a heterodimeric protein, or a heterodimeric protein substantially homologous thereto as herein defined, wherein CSR::T1R2-a corresponds to SEQ ID NO: 2, CSR::T1 R2-b corresponds to SEQ ID NO:20, CSR::T1R3-a corresponds to SEQ ID NO
  • nucleic acid encoding a CSR::T1R chimeric protein able to bind at least one sweetener or sweetness enhancer comprising one or more of a nucleic acid substantially homologous to a nucleotide sequence selected from the group consisting of SEQ ID NO:1 (CSR::T1R2-a), SEQ ID NO: 19 (CSR::T1R2-b); SEQ ID NO:3 (CSR::T1R3-a), and SEQ ID NO: 21 (CSR::T1R3-b), as determined by sequence identity, a nucleic acid substantially homologous to a nucleotide sequence selected from the group consisting of SEQ ID NO:1.
  • CSR::T1R2-a SEQ ID NO: 19 (CSR::T1R2-b); SEQ ID NO:3 (CSR::T1R3-a), and SEQ ID NO: 21 (CSR::T1R3-b), as determined by hybridisation, a nucleic acid substantially homologous to a nucleotide sequence encoding the CSR::T1R chimeric protein as defined in claim 1, wherein the substantially homologous nucleic acid as determined by sequence identity has a sequence identity of at least 90%; wherein the substantially homologous nucleic acid as determined by hybridisation hybridises under stringent hybridization conditions at a temperature of 42° C in a solution consisting of 50% formamide, 5 ⁇ SSC, and 1% SDS, and washing at 65° C in a solution consisting of 0.2 ⁇ SSC and 0.1% SDS; wherein the nucleic acid optionally comprises SEQ ID NO:6 (HSV tag) at or near its end to form the C-terminus in the corresponding protein.
  • an expression vector comprising the nucleic acid as defined herein-above.
  • a host cell transfected with an expression vector as defined in herein-above.
  • a host cell as described herein-above stably expressing a CSR::T1R chimeric protein as defined herein-above and a G-Protein, optionally a G-Protein substantially homologous to Gaq-Gustducin.
  • a host cell as described herein-above, transiently expressing a CSR::T1R chimeric protein as described herein-above and a G-Protein, optionally a G-Protein substantially homologous to Gaq-Gustducin.
  • a method of producing a CSR::T1R chimeric protein as defined herein-above comprising the step of culturing host cells comprising an expression vector encoding for the CSR::T1R chimeric protein, under conditions sufficient for expression, thereby forming the CSR::T1 R chimeric protein and optionally recovering it from the cells.
  • a method to identify an agent that modulates sweet taste signaling in taste cells comprising:
  • G-Protein is a chimeric G-protein substantially homologous to Gaq-Gustducin.
  • G-Protein is the chimeric G-protein G alpha 16-gustducin 44.
  • step (ii) is performed by measuring a change in or caused by intracellular messengers.
  • a method as defined herein-above wherein said cells are selected from the group consisting of bacterial cells, eucaryotic cells, yeast cells, insect cells, mammalian cells, amphibian cells, and worm cells.
  • a method as described herein-above wherein the cell is a mammalian cell.
  • the cell is a mammalian cell selected from the group consisting of CHO, COS, HeLa and HEK-293 cells.
  • step (i) further comprises contacting the CSR::T1 R chimeric protein with a test agent in presence of calcium.
  • kits comprising
  • kit as defined herein-above comprising:
  • a method to identify an agent that modulates the CSR::T1 R chimeric protein as defined herein-above comprising:
  • ligand is selected from the group consisting of calcium, calcium ions and calcium chloride.
  • step (i) is performed by a method selected from the group consisting of fluorescence spectroscopy, NMR spectroscopy, measuring of one or more of absorbance, refractive index, hydrodynamic methods, chromatography, measuring solubility, biochemical methods, wherein the methods measure the properties of the CSR::T1 R chimeric protein in a suitable environment selected form the group consisting of solution, bilayer membrane, attached to a solid phase, in a lipid monolayer, bound on a membrane, and in vesicles.
  • CSR::T1 R chimeric proteins include but are not limited to a CSR::T1 R2 monomer, a CSR::T1R3 monomer, a CSR::T1R2/CSR::T1R3 heterodimer, a CSR::T1R2/T1R3 heterodimer (chimeric T1 R2 subunit with wildtype T1 R3), and a T1 R2/CSR::T1R3 heterodimer (chimeric T1 R3 subunit with wildtype T1 R2).
  • CSR::T1R2 includes but is not limited to CSR::T1R2-a and CSR::T1R2-b.
  • CSR::T1R3 includes but is not limited to CSR::T1R3-a or CSR::T1R3-b.
  • Each -a variant differs from the relevant -b variant in the exact location where the two parts of different origin (CSR and T1 R, respectively) are joined to give the chimeric cSR::T1 R protein.
  • variants-a are joined just before the cysteine-rich domain (CRD), variants-b are joined just after the CRD, as is apparent from their sequences (SEQ ID NO: 1 + 2: CSR::T1 R2-a nucleic acid + protein; SEQ ID NO: 3 + 4: CSR::T1 R3-a nucleic acid + protein; SEQ ID NO: 19 + 20: CSR::T1 R2-b nucleic acid + protein; SEQ ID NO: 21 + 22: CSR::T1 R3-b nucleic acid + protein).
  • the CSR::T1 R chimeric proteins include, in particular, a CSR::T1 R2-a monomer, a CSR::T1 R3-a monomer, a CSR::T1 R2-a/CSR::T1 R3-a heterodimer, a CSR::T1R2-a/T1R3 heterodimer (chimeric T1R2-a subunit with wildtype T1R3), and a T1R2/CSR::T1R3-a heterodimer (chimeric T1R3-a subunit with wildtype T1 R2), as well as a CSR::T1 R2-b monomer, a CSR::T1 R3-b monomer, a CSR:T1R2-b/CSR::T1R3-b heterodimer, a CSR:T1 R2-b/T1 R3 heterodimer, a CSR:T1 R2-b/T1 R3 heterodimer, a CSR:
  • the CSR:T1 R chimeric protein does not possess the VFT domains of T1 R2, T1 R3, or T1 R2 and T1R3, and therefore allows specifically identification of compounds that bind to the TMD domains and the cystein-rich domains of T1 R2 and/or T1 R3. These identified compounds are of particular interest as they would not be expected to compete with carbohydrates binding in the VFT site for binding to the sweet taste receptor in vivo and are therefore particularly interesting potential candidates for sweetness enhancers of carbohydrates.
  • Chimeric proteins are joined fragments of two or more original proteins that sometimes are able to combine desired properties or eliminate unwanted ones.
  • folding of a protein in the three dimensional space is critical and the position of amino acids will influence the folding, not any two fragments can be joined. Even if critical domains and amino acids are known, the successful expression, correct folding and intact functionality of desired properties is very much unpredictable.
  • CSR::T1R2 and CSR::T1 R3 are functional and are able to form a functional CSR::T1R2/CSR::T1R3 heterodimer (see examples for the CSR::T1 R2-a/CSR::T1 R3-a and CSR::T1 R2-b/CSR::T1 R3-b heterodimers; CSR::T1 R2-a/CSR::T1 R3-b and CSR::T1 R2-b/CSR::T1 R3-a heterodimers may also work).
  • CSR::T1R2 monomeric subunit also functions as a functional sweet receptor on its own, without forming a heterodimer.
  • Preliminary experiments indicate that the CSR::T1R3 may have difficulties in engaging and/or activating certain G-proteins; however, CSR::T1R3 is useful in binding assays that do not require the ability to activate a G-Protein.
  • CSR::T1R2 and CSR::T1R3 are also useful in their monomeric form in the methods described herein.
  • Alternative heterodimers that can be used in these methods are chimeric subunit/wildtype subunit heterodimers (CSR::T1R2/T1R3 and T1R2/CSR::T1R3).
  • each of the CSR::T1 R subunits of the heterodimeric complex consist of joined sequence fragments from two source proteins.
  • the two source proteins are the human calcium-sensing receptor (hCaSR), and a T1R protein (T1R2 or T1R3).
  • the hCaSR-derived fragment (CSR) common to both subunits comprises the extracellular domain (ECD) of hCaSR.
  • the T1R-derived fragments comprise the transmembrane domains (TMD) of the T1 R sequences and differ, as they are derived from either T1 R2 or T1 R3.
  • the chimeric protein allows to use calcium as a ligand/agonist for receptor activation instead of sweeteners, so that any adverse effects due to the presence of sweeteners can be avoided.
  • CSR::T1R designates the CSR::T1R2 homomer; or the CSR::T1R3 homomer; or the heterodimeric complex of CSR::T1R2 with CSR::T1R3 or with the wildtype T1 R3 (CSR::T1 R2/CSR::T1 R3 or CSR::T1 R2/T1 R3); or the heterodimeric complex of CSR::T1R3 with CSR::T1R2 or with the wildtype T1 R2 (CSR::T1 R2/CSR::T1 R3 or T1 R2/CSR::T1 R3).
  • CSR::T1R is also referred to as "the GPCR”.
  • the chimeric CSR::T1R constructs that are provided are useful when screening, without limitation, for modulators of the sweet taste response, for example, without limitation, sweetness enhancers.
  • Traditional screening methods and binding assays may be used to screen for modulators and enhancers. Such screening methodology is well-known in the art, and is outlined below.
  • the CSR (part of the hCaSR) in the chimeric CSR::T1R constructs renders the resulting receptors responsive to calcium
  • the ligand is replaced with calcium (for example, without limitation, in the form of calcium chloride).
  • This has the additional advantage of avoiding any negative effects of the actual ligand/agonist being present. For example, when screening for modulators of sugar ligands/agonists, the adverse effects of sugars on osmolarity etc. is avoided.
  • Transfected or endogenous T1 R3 and T1 R2 can negatively interfere with methods that determine agonist responses of CSR::T1R2 and/or CSR::T1R3, respectively, or the change of said responses dependent on another modulator.
  • the absence of T1 R3 and T1 R2 provides a null background for the determination of CSR::T1R2 and/or CSR::T1 R3 activation, so that observed signals can be directly attributed to CSR::T1 R2 and/or CSR::T1 R3 activity.
  • T1 R3 is part of both the sweet and the umami heterodimers.
  • T1 R2 and/or T1 R3 will cause some backgrounds signals, which are undesirable. While cells with endogenous T1R2 and/or T1 R3 can still be useful to obtain results with sufficiently low background, a better choice are cells that do not contain the endogenous T1 R2 and T1 R3 receptors.
  • An exception occurs when using a CSR::T1 R2/T1 R3 chimeric protein, which may contain wildtype T1 R3 without adverse effect on the background, or a T1 R2/CSR::Tl R3 chimeric protein, which may contain wildtype T1 R2 without adverse effects on the background.
  • the cells listed below are particularly useful as they do not contain endogenous/wildtype T1R3, or endogenous wildtype T1R2.
  • Suitable eucaryotic cells include eucaryotic cells, for example, without limitation, mammalian cells, yeast cells, or insect cells (including Sf9), amphibian cells (including melanophore cells), or worm cells including cells of Caenorhabditis (including Caenorhabditis elegans).
  • Suitable mammalian cells include, for example, without limitation, COS cells (including Cos-1 and Cos-7), CHO cells, HEK293 cells, HEK293T cells, HEK293 T-RexTM cells, or other transfectable eucaryotic cell lines.
  • Suitable bacterial cells include without limitation E. coli.
  • Cells may be transfected with a GPCR and a G-protein (which links the receptor to a phospholipase C signal transduction pathway) transiently or stably, as is well known in the art.
  • G-protein G alpha 16-gustducin 44 also known as G.sub..alpha.16 gust(ducin)44, G.sub.alpha.16gust(ducin)44, G ⁇ 16gust(ducin)44, Ga16gust(ducin)44, G ⁇ 16-gustducin 44, or as used herein-below, "G16gust44”
  • G16gust44 chimeric G-proteins based on Gaq-Gustducin described in WO 2004/055048 , or other G-Proteins, for example, G16 or G15, may also be used.
  • the CSR::T1R can be expressed in a cell with a G-protein that links the receptor to a signal transduction pathway, for example, the phospholipase C signal transduction pathway, or signal transduction pathways including, for example, the following: adenylate cyclase, guanylate cyclase, phospholipase C, IP3, GTPase/GTP binding, arachinoid acid, cAMP/cGMP, DAG, protein kinase c (PKC), MAP kinase tyrosine kinase, or ERK kinase.
  • a signal transduction pathway for example, the phospholipase C signal transduction pathway, or signal transduction pathways including, for example, the following: adenylate cyclase, guanylate cyclase, phospholipase C, IP3, GTPase/GTP binding, arachinoid acid, cAMP/cGMP, DAG
  • any suitable reporter gene may be linked to a CSR::T1 R-activation responsive promoter and used to determine CSR::T1R activity, as described in more detail herein-below.
  • the vector constructs for expressing the GPCR and/or the G-protein in such cells may be produced in a manner known per se using Polymerase Chain Reactions. After verfication of the sequence, cDNA fragments may be sub-cloned into a suitable vector, for example pcDNA 3.1 mammalian expression vector for mammalian cells, and transiently transfected in a corresponding host cell to enable the correct expression of the gene.
  • a suitable vector for example pcDNA 3.1 mammalian expression vector for mammalian cells, and transiently transfected in a corresponding host cell to enable the correct expression of the gene.
  • cell lysates may be prepared, analysed by a Western-Blot analysis in order to confirm the correct expression of the protein.
  • suitable cells for example mammalian cells including HEK293T cells and HEK T-Rex TM , may be transfected to generate cells stably expressing the protein according to techniques well known in the art.
  • non-mammalian expression vector/host systems can be used to contain and express sequences encoding the CSR::T1R G-Protein coupled receptor (GPCR).
  • GPCR G-Protein coupled receptor
  • microorganisms including bacteria transformed with recombinant bacteriophage, plasmid, or cosmid DNA expression vectors; yeast transformed with yeast expression vectors; insect cell systems infected with viral expression vectors (for example baculovirus), or with bacterial expression vectors (for example pBR322 plasmids).
  • cloning and expression vectors may be selected depending upon the use intended for polynucleotide sequences encoding the GPCR. For example, routine cloning, subcloning, and propagation of polynucleotide sequences encoding a GPCR can be achieved using a multifunctional E. coli vector such as pBLUESCRIPT (Stratagene, La Jolla Calif.) or pSPORT1 plasmid (Life Technologies). Ligation of sequences encoding a GPCR into the vector's multiple cloning site disrupts the lacZ gene, allowing a colorimetric screening procedure for identification of transformed bacteria containing recombinant molecules.
  • pBLUESCRIPT Stratagene, La Jolla Calif.
  • pSPORT1 plasmid Life Technologies
  • these vectors may be useful for in vitro transcription, dideoxy sequencing, single strand rescue with helper phage, and creation of nested deletions in the cloned sequence.
  • vectors which direct high level expression of a GPCR may be used.
  • vectors containing the strong, inducible SP6 or T7 bacteriophage promoter may be used.
  • Yeast expression systems may be used for production of a GPCR.
  • a number of vectors containing constitutive or inducible promoters, such as alpha factor, alcohol oxidase, and PGH. promoters, may be used in the yeast Saccharomyces cerevisiae or Pichia pastoris.
  • such vectors direct either the secretion or intracellular retention of expressed proteins and enable integration of foreign sequences into the host genome for stable propagation.
  • heterologous proteins in insect cell lines is, for example, derivatives of the Lepidopteran baculovirus, Autographa californica multicapsid nucleo-virus (A c MNPV) can be used.
  • a c MNPV Autographa californica multicapsid nucleo-virus
  • foreign gene expression is directed by a very strong late viral promoter, either the polyhedrin or p10 promoters, and a wide array of vectors is available that optimises expression and recovery of recombinant proteins.
  • These vectors enable expression of both membrane-bound and secreted proteins at high levels, and also many post-translational modifications known to occur in mammalian systems, including N- and O -linked glycosylation, phosphorylation, acylation, proteolysis and secreted vaccine components.
  • a number of vectors are commercially available, for example the InsectSelectTM System from Invitrogen.
  • cDNAs encoding the desired proteins GPCR (CSR::T1 R) and G-protein
  • CSR CSR::T1 R
  • G-protein G-protein
  • a strong promoter to direct transcription a transcription/translation terminator
  • a ribosome-binding site for translational initiation Suitable bacterial promoters are well known in the art, for example, E. coli, Bacillus sp., and Salmonella, and kits for such expression systems are commercially available.
  • eukaryotic expression systems for mammalian cells, yeast, and insect cells are commercially available.
  • the eukaryotic expression vector may be, for example, an adenoviral vector, an adeno-associated vector, or a retroviral vector.
  • the expression vector typically contains a transcription unit or expression cassette that contains all the additional elements required for the expression of the protein-encoding nucleic acid in host cells.
  • a typical expression cassette thus contains a promoter operably linked to the nucleic acid sequence encoding the protein and signals required for efficient polyadenylation of the transcript, ribosome binding sites, and translation termination.
  • the nucleic acid sequence encoding the protein may typically be linked to a membrane-targeting signal such as the N-terminal 45 amino acids of the rat Somatostatin-3 receptor sequence to promote efficient cell-surface expression of the recombinant protein, which is useful for cell-surface receptors. Additional elements may include, for example, enhancers.
  • An expression cassette should also contain a transcription termination region downstream of the structural gene to provide for efficient termination.
  • the termination region may be obtained from the same gene as the promoter sequence or may be obtained from different genes.
  • vectors for expression in eucaryotic or procaryotic cells well known in the art may be used.
  • vectors include bacterial expression vectors, for example, plasmids including pBR322-based plasmids, pSKF, and pET23D, and fusion expression systems , for example, GST and LacZ.
  • Expression vectors containing regulatory elements from eukaryotic viruses are typically used in eukaryotic expression vectors, for example SV40 vectors, cytomegalovirus vectors, papilloma virus vectors, and vectors derived from Epstein-Barr virus.
  • exemplary eukaryotic vectors include pMSG, pAV009/A + , pMT010/A + , pMAMneo-5, baculovirus pDSVE, pcDNA3.1, pIRES and any other vector allowing expression of proteins under the direction of the SV40 early promoter, SV40 later promoter, metallothionein promoter, murine mammary tumor virus promoter, Rous sarcoma virus promoter, polyhedrin promoter, or other promoters shown effective for expression in eukaryotic cells.
  • Some expression systems have markers that provide gene amplification such as thymidine kinase, hygromycin B phosphotransferase, dihydrofolate reductase and the like.
  • the elements that are typically included in expression vectors may also include a replicon that functions in E. coli, a gene encoding drug resistance to permit selection of bacteria that harbor recombinant plasmids, and unique restriction sites in non-essential regions of the plasmid to allow insertion of eukaryotic sequences.
  • the particular drug resistance gene chosen is not critical, any of the many drug resistance genes known in the art are suitable.
  • the prokaryotic sequences are optionally chosen such that they do not interfere with the replication of the DNA in eukaryotic cells, if necessary.
  • the GPCR cDNA fragment may be expressed alone or as a fusion protein wherein the GPCR of interest is fused to the E. coli periplasmic maltose-binding protein (MBP) wherein the MBP, including its signal peptide, is linked to the amino terminus of the GPCR.
  • MBP E. coli periplasmic maltose-binding protein
  • the wild-type GPCR cDNA or the MBP:GPCR fusion cDNA is subcloned into a suitable plasmid, for example pBR322, where in E. coli, GPCR expression is driven by the lac wild-type promoter.
  • coli are described, for example, in " G-protein coupled receptors (Signal Transduction Series)"; Editors: Tatsuya Haga and Gabriel Berstein, 1st ed., pp. 265-280 CRC Press - Boca Raton FL; September 1999 .
  • Genetically engineered yeast systems and insect cell systems which lack endogenous GPCRs provide the advantage of a null background for CSR::T1 R activation screening.
  • yeast systems substitute a human GPCR and Ga protein for the corresponding components of the endogenous yeast pheromone receptor pathway. Downstream signaling pathways are also modified so that the normal yeast response to the signal is converted to positive growth on selective media or to reporter gene expression (described by Broach, J. R. and J. Thorner (1996) Nature 384 (supp.):14-16 ).
  • Amphibian cell systems in particular melanophore cells, are described, for example, in WO 92/01810 that describes a GPCR expression system.
  • CSR::T1R may be overexpressed by placing it under the control of a strong constitutive promoter, for example the CMV early promoter.
  • a strong constitutive promoter for example the CMV early promoter.
  • certain mutations of conserved GPCR amino acids or amino acid domains can be introduced to render the employed GPCR constitutively active.
  • Standard transfection methods can be used to produce bacterial, mammalian, yeast or insect cell lines that express large quantities of the protein.
  • Any known method for introducing nucleotide sequences into host cells may be used. It is only necessary that the particular genetic engineering procedure used be capable of successfully introducing the relevant genes into the host cell capable of expressing the proteins of interest. These methods may involve introducing cloned genomic DNA, cDNA, synthetic DNA or other foreign genetic material into a host cell and include the use of calcium phosphate transfection, polybrene, protoplast fusion, electroporation, liposomes, microinjection, plasma vectors, viral vectors and the like. For example, without limitation, the T-Rex TM expression system (Invitrogen Corp., Carlsbad, CA) may be used.
  • the T-Rex TM System is a tetracycline-regulated mammalian expression system that uses regulatory elements from the E. coli Tn10-encoded tetracycline (Tet) resistance operon. Tetracycline regulation in the T-Rex TM System is based on the binding of tetracycline to the Tet repressor and derepression of the promoter controlling expression of the gene of interest.
  • the transfected cells may be cultured using standard culturing conditions well known in the art. It will be apparent to the skilled person that different cells require different culture conditions including approriate temperature and cell culture media.
  • the protein may be recovered from the cell culture using standard techniques.
  • the cells may be burst open either mechanically or by osmotic shock before being subject to precipitation and chromatography steps, the nature and sequence of which will depend on the particular recombinant material to be recovered.
  • the recombinant protein may be recovered from the culture medium in which the recombinant cells had been cultured.
  • Modulators (various types including ligands, agonists, partial agonists, antagonists, inverse agonists, inhibitors, enhancers) of CSR::T1R receptor activity can be identified as described herein below.
  • the type of a modulator may include more than one type at a time, and may depend on the concentration. For example, an agent may act as an agonist in a certain concentration range, but act as a modulator or enhancer of another agonist (for example a sweetener or sugar) in another concentration range. Therefore, agents should be tested at different concentrations to identify them as modulators.
  • a modulator is an agent that effects an increase or decrease of one or more of the following: the cell surface expression of a receptor, the binding of a ligand to a receptor, the intracellular response initiated by an active form of the receptor (either in the presence or absence or an agonist).
  • the modulator can itself be an agonist that binds to the receptor, activates it and thereby modulates an increase in the cellular response.
  • Modulators include various types of compounds, including small molecules, peptides, proteins, nucleic acids, antibodies or fragments thereof. These can be derived from various sources including synthetic or natural, extracts of natural material, for example from animal, mammalian, insect, plant, bacterial or fungal cell material or cultured cells, or conditioned medium of such cells.
  • a ligand is an agent that binds to the receptor; it may be an agonist, partial agonist, enhancer, antagonist, or inverse agonist.
  • An agonist is a ligand of the CSR::T1 R chimeric protein receptor that activates the receptor and increases an intracellular response when it binds to a receptor compared to the intracellular response in the absence of the agonist. Additionally or alternatively, an agonist may decrease internalization of a cell surface receptor such that the cell surface expression of a receptor is increased as compared to the number of cell surface receptors present on the surface of a cell in the absence of an agonist.
  • Agonists of CSR::T1 R include, for example, calcium, perillartine, cyclamate, NDHC, and cinnamonitrile.
  • a ligand of the CSR::T1R chimeric protein can be divided into two types, a CSR-domain-ligand which binds in the CSR part of the chimeric protein (calcium), or a TSR-domain ligand, which binds in the T1 R-part of the chimeric protein (modulators of the sweet taste response).
  • a partial agonist is an agonist that only partially activates the receptor in comparison to other agonists that maximally activate the receptor.
  • An antagonist is a ligand which binds to the receptor at the same (competitive antagonist) or at a different site (alllosteric antagonist) as an agonist, but does not activate an intracellular response initiated by an active form of a receptor, thereby inhibiting the intracellular response induced by an agonist as compared to the intracellular response in the presence of an agonist and in the absence of an antagonist.
  • An inverse agonist binding to a receptor, decreases the constitutive intracellular response mediated by a receptor as compared to the intracellular response in the absence of the inverse agonist.
  • An inhibitor decreases the binding of an agonist to the receptor as compared to the binding of the agonist in the absence of inhibitor, and/or decreases the intracellular response induced by an agonist.
  • An enhancer increases the binding of an agonist to the receptor as compared to the binding of the agonist in the absence of enhancer, and/or increases the intracellular response induced by an agonist.
  • the activity, or changes in activity, of a receptor binding a ligand and transmitting the signal through, for example, a G-protein can be determined by the assays described herein-below.
  • Modulators can be identified using a wide variety of in vitro and in vivo assays to determine and compare functional effects/parameters, or alternatively by binding assays.
  • the effects of the test agents upon the function of the receptors can be measured by examining a suitable functional parameters. Any physiological change that affects receptor activity can be used to identify modulators.
  • Such functional assays are well-known in the art, for example assays using intact cells or tissues isolated from animals based on measuring the concentration or activity or their change of a secondary messenger (including, for example, intracellular calcium (Ca2+), cAMP, cGMP, inositol phospate (IP3), diacylglycerol/DAG, arachinoid acid, MAP kinase or tyrosine kinase), ion flux, phosphorylation levels, transcription levels, neurotransmitter levels, and assays based on GTP-binding, GTPase, adenylate cyclase, phospholipid-breakdown, diacylglycerol, inositol triphosphate, arachidonic acid release, PKC, kinase and transcriptional reporters.
  • a secondary messenger including, for example, intracellular calcium (Ca2+), cAMP, cGMP, inositol phospate (IP
  • Receptor activation typically initiates subsequent intracellular events, for example, increases in second messengers, for example, IP3, which releases intracellular stores of calcium ions.
  • IP3 inositol triphosphate
  • IP3 phospholipase C-mediated hydrolysis of phosphatidylinositol.
  • IP3 in turn stimulates the release of intracellular calcium ion stores.
  • a change in cytoplasmic calcium ion levels, or a change in second messenger levels such as IP3 can be used to determine G-protein coupled receptor activity.
  • All functional assays may be performed by samples containing cells expressing the receptor on their surfaces or on isolated cell membrane fractions. Useful cells are described herein-above. Instead of samples with separate cells or cell membranes, tissues from transgenic animals may be used.
  • the screening methods described herein are particularly useful to identify modulators of the sweet taste response, for example, sweetness enhancers.
  • a modulator which is not an agonist itself e.g. an antagonist, partial agonist, inverse agonist, inhibitor, or enhancer
  • samples with and without test agent both containing an agonist are compared.
  • agonist for example, calcium can be used. Using calcium has the advantage that both TMDs will be accessible.
  • Other known or identified agonists can also be used, for example, perillartine, cyclamate, neohesperidine dihydrochalone (NDHC), and cinnamonitrile), but will partially occupy ligand/agonist binding sites which may coincide with the modulator binding site of the to be identified modulator, and may cause lower signals.
  • a control (with agonist but without modulator) is assigned a relative receptor activity value of 100.
  • a decrease in activity relative to the control identifies an inhibitor, antagonist or inverse agonist, an increase identifies an enhancer.
  • an increase or decrease in the measured activity of 10% or more in a sample with test agent compared to a sample without test agent or compared to a sample with test agent but based on cells that do not express CSR::T1 R (mock-transfected cells) can be considered significant.
  • samples with and without test agent are compared.
  • a control with agonist, for example calcium chloride, but without modulator
  • a relative receptor activity value of 100 is assigned.
  • An increase identifies an enhancer.
  • an increase or decrease in the measured activity of 10% or more in a sample with test agent compared to a sample without test agent or compared to a sample with test agent but based on cells that do not express CSR::T1R (mock-transfected cells) can be considered significant.
  • CSR::TSR1 chimeric protein calcium can be used as agonist.
  • agonists binding in the relevant parts of the T1 R2 and/or T1 R3 fragments of CSR::TSR1 may be used.
  • These agonists include, for example, perillartine, cyclamate, NDHC, and cinnamonitrile.
  • a sample with test agent is compared to a positive control with an agonist (for example calcium chloride, perillartine, cyclamate, neohesperidin dihydrochalcone (NDHC), cinnamonitrile, or another identified ligand/agonist).
  • an agonist for example calcium chloride, perillartine, cyclamate, neohesperidin dihydrochalcone (NDHC), cinnamonitrile, or another identified ligand/agonist.
  • samples with and without test agent are compared in their activity of the CSR::T1R chimeric protein.
  • an agonist or partial agonist will have a biological activity corresponding to at least 10% of the maximal biological activity of the positive control sweet agonist when the agonist or partial agonist is present at 100 mM or less, for example it may have a maximal biologial activity comparable to the agonist or higher.
  • Maximal biological activity is defined as the maximal achievable receptor response to an agonist, for example calcium chloride, perillartine, cyclamate, neohesperidin dihydrochalcone (NDHC), cinnamonitrile that can be achieved within a given receptor assay format and this response fails to increase further despite application of increasing concentrations of that same agonist.
  • the above-mentioned agonists may, at a different concentration, also act as an enhancer of an agonist of the CSR::T1R chimeric protein. This may be tested in a screening method by using calcium or other agonist to test the agonist-test agent for signals indicating a sweetness enhancing effect.
  • an increase in the measured activity of, for example, 10% or more in a sample with test agent is compared to a sample without test agent or is compared to a sample with test agent but based on cells that do not express CSR::T1 R (mock-transfected cells).
  • Antagonists show a reduction of agonist-stimulated receptor activity, for example by at least 10%.
  • receptor activity in the presence of a known agonist with and without a test agent is compared in samples comprising animals/cells/membranes that overexpress the receptor as described herein-above.
  • Inverse agonists show a reduction of constitutive activity of the receptor, for example by at least 10%.
  • Intracellular calcium release induced by the activation of GPCRs is detected using cell-permeant dyes that bind to calcium.
  • the calcium-bound dyes generate a fluorescence signal that is proportional to the rise in intracellular calcium. The methods allows for rapid and quantitative measurement of receptor activity.
  • Cells used are transfected cells that co-express the CSR::T1R GPCR and a G-protein which allows for coupling to the phospholipase C pathway as described herein-above.
  • Negative controls include cells or their membranes not expressing CSR::T1 R (mock transfected), to exclude possible non-specific effects of the candidate compound.
  • the calcium flux detection protocol is described in detail in " G-protein coupled receptors (Signal Transduction Series)"; Editors: Tatsuya Haga and Gabriel Berstein, 1st ed., 424pp.CRC Press - Boca Raton FL; September 1999 , and an adapted version with is summarised below:
  • Useful cells are, without limitation, mammalian cells as described herein-above, for example HEK293T cells and HEK293 T-RexTM cells.
  • Cells may be transfected with a GPCR and a G-Protein transiently or stably as is well known in the art.
  • An excellent heterologous expression system is described in detail in WO 2004/055048 .
  • a calcium flux assay can be performed, for example, as described in example 1 herein-below.
  • the identification of a modulator is performed as described above subject to the following modifications.
  • the signals are compared to the baseline level of CSR::T1 R activity obtained from recombinant cells expressing CSR::T1R in the presence of an agonist but in the absence of a test agent.
  • An increase or decrease in CSR::T1 R activity for example of at least 2 fold, at least 5 fold, at least 10 fold , at least a 100 fold, or more identifies a modulator.
  • the identification involves an increase or decrease fluorescence intensity of, for example, 10% or more, when compared to a sample without modulator, or when compared to a sample with modulator but in cells that do not express the CSR::T1 R polypeptide (mock-transfected cells).
  • Assays for adenylate cyclase activity are performed, for example, as described in detail by Kenimer & Nirenberg, 1981, Mol. Pharmacol. 20: 585-591 . Reaction mixtures are incubated usually at 37° C for less than 10 minutes. Following incubation, reaction mixtures are deproteinized by the addition of 0.9 ml of cold 6% trichloroacetic acid. Tubes are centrifuged and each supernatant solution is added to a Dowex AG50W-X4 column.
  • the cAMP fraction from the column is eluted with 4 ml of 0.1 mM imidazole-HCl (pH 7.5) into a counting vial in order to measure the levels of cAMP generated following receptor activation by the agonist.
  • Control reactions should also be performed using protein homogenate from cells that do not express a CSR::T1 R polypeptide.
  • inositol triphosphate (IP3)/Ca 2+ and thereby receptor activity can be detected using fluorescence.
  • Cells expressing a GPCR may exhibit increased cytoplasmic calcium levels as a result of contribution from both intracellular stores and via activation of ion channels, in which case it may be desirable, although not necessary, to conduct such assays in calcium-free buffer, optionally supplemented with a chelating agent such as EDTA, to distinguish fluorescence response resulting from calcium release from internal stores.
  • a chelating agent such as EDTA
  • CSR::T1 R is expressed in a cell with a G-protein that links the receptor to a phospholipase C signal transduction pathway. Changes in intracellular Ca 2+ concentration are measured, for example using fluorescent Ca 2+ indicator dyes and/or fluorometric imaging.
  • a measure of receptor activity is the binding of GTP by cell membranes containing the GPCR. Measured is the G-protein coupling to membranes by detecting the binding of labelled GTP.
  • Membranes isolated from cells expressing the receptor are incubated in a buffer containing 35S-GTP ⁇ S and unlabelled GDP. Active GTPase releases the label as inorganic phosphate, which is detected by separation of free inorganic phosphate in a 5% suspension of activated charcoal in 20 mM H 3 PO 4 , followed by scintillation counting. The mixture is incubated and unbound labelled GTP is removed by filtration onto GF/B filters.
  • Bound and labelled GTP is measured by liquid scintillation counting.
  • Controls include assays using membranes isolated from cells not expressing CSR::T1 R (mock-transfected), in order to exclude possible non-specific effects of the test agent. The method is described in detail by Traynor and Nahorski, 1995, Mol. Pharmacol. 47: 848-854 .
  • an agent is identified as an agonist usually if the activity is at least 50% of that of a known agonist (for example perillartine) when the compound is present at 100 mM or less, for example 10 to 500 ⁇ M, for example about 100 ⁇ M, or if it will induce a level the same as or higher than that induced by a known agonist.
  • a known agonist for example perillartine
  • Microphysiometer or biosensor
  • Such assays can be performed as described in detail in Hafner, 2000, Biosens. Bioelectron. 15: 149-158 .
  • the intracellular level of arachinoid acid is employed as an indicator of receptor activity. Such a method is described in detail by Gijon et al., 2000,J. Biol. Chem., 275:20146-20156 .
  • Intracellular or extracellular cAMP is measured using a cAMP radioimmunoassay (RIA) or cAMP binding protein, for example as described by Horton & Baxendale, 1995, Methods Mol. Biol. 41: 91-105 .
  • a number of kits for the measurement of cAMP are commercially available, for example the High Efficiency Fluorescence Polarization-based homogeneous assay by LJL Biosystems and NEN Life Science Products.
  • the intracellular or extracellular levels of cGMP may measured using an immunoassay.
  • the method described in Felley-Bosco et al., Am. J. Resp. Cell and Mol. Biol., 11:159-164 (1994 ) may be used to determine the level of cGMP.
  • an assay kit for measuring cAMP and/or cGMP as described in US 4,115,538 can be used.
  • Negative controls with mock-transfected cells or extracts thereof to exclude possible non-specific effects of test agents may be used.
  • DAG Diacylglycerol
  • IP3 inositol triphosphate
  • CSR::T1 R GPCR
  • Negative controls with mock-transfected cells or extracts thereof to exclude possible non-specific effects of test agents may be used.
  • PKC Protein Kinase C
  • Gene products induced by PKC show PKC activation and thereby receptor activity.
  • gene products include, for example, proto-oncogene transcription factor-encoding genes (including c-fos, c-myc and c-jun), proteases, protease inhibitors (including collagenase type I and plasminogen activator inhibitor), and adhesion molecules (including intracellular adhesion molecule I (ICAM I)).
  • proto-oncogene transcription factor-encoding genes including c-fos, c-myc and c-jun
  • proteases including collagenase type I and plasminogen activator inhibitor
  • adhesion molecules including intracellular adhesion molecule I (ICAM I)
  • PKC activity may be directly measured as described by Kikkawa et al., 1982, J. Biol. Chem. 257: 13341 , where the phosphorylation of a PKC substrate peptide, which is subsequently separated by binding to phosphocellulose paper, is measured. It can be used to measure activity of purified kinase, or in crude cellular extracts. Protein kinase C sample can be diluted in 20 mM HEPES/ 2 mM DTT immediately prior to the assay.
  • activity can be measured through the use of reporter gene constructs driven by the control sequences of genes activated by PKC activation.
  • Negative controls with mock-transfected cells or extracts thereof to exclude possible non-specific effects of test agents may be used.
  • MAP kinase activity can be measured using commercially available kits, for example, the p38 MAP Kinase assay kit by New England Biolabs, or the FlashPlateTM MAP Kinase assays by Perkin-Elmer Life Sciences.
  • p42/44 MAP kinases or ERK1/2 can be measured to show GPCR (CSR::T1R) activity when cells with Gq and Gi coupled GPCRs are used, and an ERK1/2 assay kit is commercially available by TGR Biosciences, which measures the phosphorylation of endogenous ERK1/2 kinases following GPCR activation.
  • tyrosine kinase activity through known synthetic or natural tyrosine kinase substrates and labelled phosphate are well known; the activity of other types of kinases (for example, Serine/Threonine kinases) can be measured similarly.
  • All kinase assays can be performed with both purified kinases and crude extracts prepared from cells expressing one or more CSR::T1 R polypeptide.
  • the substrates of kinases that are used can be either full-length protein or synthetic peptides representing the substrate. Pinna & Ruzzene (1996, Biochem. Biophys. Acta 1314: 191-225 ) lists a number of phosphorylation substrate sites useful for detecting kinase activities. A number of kinase substrate peptides are commercially available.
  • RRLIEDAEYAARG commercially available from Sigma
  • RRLIEDAEYAARG a substrate for many receptor and nonreceptor tyrosine kinases.
  • Some methods require the binding of peptide substrates to filters, then the peptide substrates should have a net positive charge to facilitate binding.
  • peptide substrates should have at least 2 basic residues and a free-amino terminus. Reactions generally use a peptide concentration of 0.7-1.5 mM.
  • Negative controls with mock-transfected cells or extracts thereof to exclude possible non-specific effects of test agents may be used.
  • an at least 2-fold increase or 10% decrease in the signal is significant.
  • An agonist stimulates for example at least 2-fold, 5-fold, 10-fold or more when comparing activity in presence and absence of the test agent.
  • the intracellular signal initiated by binding of an agonist to CSR::T1R sets in motion a cascade of intracellular events, the ultimate consequence of which is a rapid and detectable change in the transcription or translation of one or more genes.
  • the activity of the receptor can therefore be determined by measuring the expression of a reporter gene driven by a promoter responsive to CSR::T1 R activation.
  • a “promoter” as used herein is one or more transcriptional control elements or sequences necessary for receptor-mediated regulation of gene expression, including one or more of basal promoter, enhancers and transcription-factor binding sites necessary for receptor-regulated expression. Promoters responsive to the intracellular signals resulting from agonist binding to CSR::T1 R are selected and operatively linked to a corresponding promoter-controlled reporter gene whose transcription, translation or ultimate activity is readily detectable and measurable.
  • Reporter genes may be selected, for example, from luciferase, CAT, GFP, ⁇ -lactamase, ⁇ -galactosidase, and the so-called "immediate early" genes, c-fos proto-oncogene, transcription factor CREB, vasoactive intestinal peptide (VIP) gene, the somatostatin gene, the proenkephalin gene, the phosphoenolpyruvate carboxy-kinase (PEPCK) gene, genes responsive to NF- ⁇ B, and AP-1-responsive genes (including the genes for Fos and Jun, Fos-related antigens (Fra) 1 and 2, I ⁇ B ⁇ , ornithine decarboxylase, and annexins I and II).
  • Promoters will be selected according to the selected reporter gene, as will be apparent to the skilled person.
  • Luciferase, CAT, GFP, ⁇ -lactamase, ⁇ -galactosidase and assays for the detection of their products are well known in the art. Examples of further reporter genes are described herein-below.
  • reporter genes are suitable and are rapidly induced (for example within minutes of contact between the receptor and the effector protein or ligand).
  • Desirable properties in reporter genes include one or more of the following: rapid responsiveness to ligand binding, low or undetectable expression in quiescent cells; induction that is transient and independent of new protein synthesis; subsequent shut-off of transcription requires new protein synthesis; and mRNAs transcribed from these genes which have a short half-life of several minutes to a few hours.
  • the promoter may have one, several or all of these properties.
  • the c-fos proto-oncogene is an example of a gene that is responsive to a number of different stimuli and has an rapid induction.
  • the c-fos regulatory elements include a TATA box that is required for transcription initiation; two upstream elements for basal transcription, and an enhancer, which includes an element with dyad symmetry and which is required for induction by TPA, serum, EGF, and PMA.
  • the 20 bp c-fos transcriptional enhancer element located between -317 and -298 bp upstream from the c-fos mRNA cap site, is essential for serum induction in serum starved NIH 3T3 cells.
  • One of the two upstream elements is located at -63 to -57 and it resembles the consensus sequence for cAMP regulation.
  • the transcription factor CREB (cyclic AMP responsive element binding protein) is responsive to levels of intracellular cAMP. Therefore, the activation of a receptor that signals via modulation of cAMP levels can be determined by detecting either the binding of the transcription factor, or the expression of a reporter gene linked to a CREB-binding element (termed the CRE, or cAMP response element).
  • the DNA sequence of the CRE is TGACGTCA. Reporter constructs responsive to CREB binding activity are described in US 5,919,649 .
  • VIP vasoactive intestinal peptide
  • somatostatin gene and its promoter which is cAMP responsive
  • proenkephalin and its promoter which is responsive to cAMP, nicotinic agonists, and phorbol esters
  • PPCK phosphoenolpyruvate carboxy-kinase
  • reporter genes and their promoters that are responsive to changes in GPCR activity include the AP-1 transcription factor and NF- ⁇ B.
  • the AP-1 promoter is characterised by a consensus AP-1 binding site which is the palindrome TGA(C/G)TCA.
  • the AP-1 site is also responsible for mediating induction by tumor promoters including the phorbol ester 1 2-O-tetradecanoylphorbol-fi-acetate (TPA), and are therefore sometimes also referred to as a TRE, for TPA-response element.
  • TPA 2-O-tetradecanoylphorbol-fi-acetate
  • TRE for TPA-response element.
  • AP-1 activates numerous genes that are involved in the early response of cells to growth stimuli.
  • AP-1-responsive genes include the genes for Fos and Jun (which proteins themselves make up AP-1 activity), Fos-related antigens (Fra) 1 and 2, I ⁇ B ⁇ , ornithine decarboxylase, and annexins I and II.
  • the NF- ⁇ B promoter/binding element has the consensus sequence GGGGACTTTCC.
  • Genes responsive to NF- ⁇ B include for example those encoding IL-1 ⁇ , TNF- ⁇ , CCR5, P-selection, Fas ligand, GM-CSF and I ⁇ B ⁇ .
  • Vectors encoding NF- ⁇ B-responsive reporters are known in the art or can be readily formed using ordinary skill in the art, for example, synthetic NF- ⁇ B elements and a minimal promoter, or using the NF- ⁇ B-responsive sequences of a gene known to be subject to NF- ⁇ B regulation. Further, NF- ⁇ B responsive reporter constructs are commercially available from, for example, CLONTECH.
  • a given promoter construct can easily be tested by exposing GPCR (CSR::T1 R)-expressing cells, transfected with the construct, to an agonist (for example perillartine).
  • an increase of at least 2-fold in the expression of reporter gene in response to the agonist indicates that the reporter is suitable to measure GPCR (CSR::T1R) activity.
  • Controls for transcription assays include both cells not expressing GPCR (CSR::T1 R), but carrying the reporter construct, and cells with a promoterless reporter construct.
  • CSR::T1 R GPCR activity as shown by reporter gene activation
  • CSR::T1R GPCR specificity of the signal and determine the spectrum of their activity, thereby excluding any non-specific signals, for example non-specific signals via the reporter gene pathway.
  • IP Inositol Phosphates
  • Phosphatidyl inositol (PI) hydrolysis may be determined as described in US 5,436,128 , which involves labelling of cells with 3H-myoinositol for at least 48 hours or more. The labelled cells are contacted with a test agent for one hour, then these cells are lysed and extracted in chloroform-methanol-water. This is followed by separating the inositol phosphates by ion exchange chromatography and quantifying them by scintillation counting. For agonists, fold stimulation is determined by calculating the ratio of counts per minute (cpm) in the presence of tested agent, to cpm in the presence of buffer control. Likewise, for inhibitors, antagonists and inverse agonists, fold inhibition is determined by calculating the ratio of cpm in the presence of test agent, to cpm in the presence of buffer control (which may or may not contain an agonist).
  • ligand binding may be determined by binding assays that measure the binding of a ligand to a CSR::T1R receptor.
  • Binding assays are well known in the art and can be tested in solution, in a bilayer membrane, optionally attached to a solid phase, in a lipid monolayer, or in vesicles. Binding of a modulator to a CSR::T1R polypeptide can be determined, for example, by measuring changes in spectroscopic characteristics (for example fluorescence, absorbance, or refractive index), hydrodynamic methods (employing for example shape), chromatography, measuring solubility properties of a CSR::T1R polypeptide.
  • binding assays are biochemical and use membrane extracts from cells/tissue expressing recombinant CSR::T1R polypeptides. A binding assay may, for example, be performed as described for T1Rs by Adler et al. in US20050032158 , paragraphs [0169] to [0198].
  • the CSR::T1R chimeric protein useful in methods described herein may be selected from the group consisting of the polypeptide selected from SEQ ID NO:2 (CSR::T1R2-a), SEQ ID NO:4 (CSR::T1R3-a), SEQ ID NO: 20 (CSR::T1R2-b), SEQ ID NO: 22 (CSR::T1 R3-b), the chimeric heterodimer of SEQ ID NO: 2 and SEQ ID NO:4 (CSR::T1R2-a/ CSR::T1R3-a), the chimeric heterodimer of SEQ ID NO: 20 and SEQ ID NO: 22 (CSR::T1 R2-b/ CSR::T1R3-b), the chimeric heterodimer of SEQ ID NO: 2 and SEQ ID NO: 22 (CSR::T1R2-a/ CSR::T1R3-b), the chimeric heterodimer of SEQ ID NO: 20 and SEQ ID
  • the CSR::T1R chimeric protein may be a receptor (or nucleotide sequence to form such a CSR::T1R receptor) which is subtantially homologous and remains functional (i.e. binds to ligands and/or is activated by ligands, or encodes such a receptor).
  • a substantially homologous CSR::T1 R chimeric protein includes such proteins where the T1R2 or T1R3 part of CSR::T1 R2 and/or CSR::T1 R3 is replaced with the relevant part of an allelic variant or different species, including T1 R2 and/or T1 R3 from mouse, rat, hamster, ape, and dog.
  • substantially homologous CSR::T1 R nucleotide or polypeptide sequences may be formed by conservative mutations and/or point mutations and include any conservatively modified variant as detailed below.
  • nucleic acid sequences conservatively modified variants means nucleic acids which encode identical or essentially identical amino acid sequences (conservatively substituted amino acids, i.e. lysine switched to arginine and further examples as explained herein-below).
  • nucleic acid variations are "silent variations," which are one species of conservatively modified variations.
  • Each nucleic acid sequence which encodes a polypeptide also describes every possible silent variation of the nucleic acid. Therefore, each codon in a nucleic acid (except AUG, which is ordinarily the only codon for methionine, and TGG, which is ordinarily the only codon for tryptophan) can be modified to yield a functionally identical nucleic acid sequence that will produce an identical polypeptide. Accordingly, each silent variation of a nucleic acid which encodes a polypeptide is implicit in each given nucleic acid sequence.
  • amino acid substitutions may be introduced using known protocols of recombinant gene technology including PCR, gene cloning, site-directed mutagenesis of cDNA, transfection of host cells, and in-vitro transcription which may be used to introduce such changes to the CSR::T1 R sequence.
  • the variants can then be screened for taste-cell-specific GPCR functional activity.
  • Conservative substitution tables providing functionally similar amino acids are well known in the art.
  • one exemplary guideline to select conservative substitutions includes (original residue followed by exemplary substitution): ala/gly or ser; arg/lys; asn/gln or his; asp/glu; cys/ser; gln/asn; gly/asp; gly/ala or pro; his/asn or gln; ile/leu or val; leu/ile or val; lys/arg or gin or glu; met/leu or tyr or ile; phe/met or leu or tyr; ser/thr; thr/ser; trp/tyr; tyr/trp or phe; val/ile or leu.
  • An alternative exemplary guideline uses the following six groups, each containing amino acids that are conservative substitutions for one another: 1) Alanine (A), Serine (S), Threonine (T); 2) Aspartic acid (D), Glutamic acid (E); 3) Asparagine (N), Glutamine (Q); 4) Arginine (R), Lysine (1); 5) Isoleucine (I), Leucine (L), Methionine (M), Valine (V); and 6) Phenylalanine (F), Tyrosine (Y), Tryptophan (W).
  • Another alternative guideline is to allow for all charged amino acids as conservative substitutions for each other whether they are positive or negative.
  • individual substitutions, deletions or additions that alter, add or delete a single amino acid or a small percentage (for example up to 26%, or up to 20%, or up to 10%) of amino acids in an encoded sequence are also considered to be conservatively modified variations.
  • Substantially homologous nucleotide or polypeptide sequences have the degree of sequence identity or hybridize under certain stringent hybridization conditions as indicated below.
  • a substantially homologous nucleotide sequence has a % sequence identity of at least at least 90%, at least 95%, or at least 98%.
  • a substantially homologous polypeptide sequence has a % sequence identity of at least at least 90%, at least 95% or at least 98%.
  • BLAST Basic Local Alignment Search Tool
  • Blastn is used, using default parameters of BLAST version 2.2.1.3, including an EXPECT (statistical significance threshold for reporting matches against database sequences) of 10, and DUST filtering.
  • EXPECT statistical significance threshold for reporting matches against database sequences
  • DUST filtering is used.
  • Nucleotide sequences are considered substantially homologous provided that they are capable of selectively hybridizing to the nucleotide sequences presented herein, or to their complement, under stringent hybrdisation conditions detailed below.
  • Background hybridization may occur because of other nucleotide sequences present, for example, in the cDNA or genomic DNA library being screened.
  • a signal that is less than 10 fold as intense as the specific interaction observed with the target DNA is considered background.
  • the intensity of interaction may be measured, for example, by radiolabelling the probe, e.g. with 32P.
  • a kit for example, a screening kit or high throughput screening kit, that comprises recombinant cells that express the CSR::T1R, or a substantially homologous sequence thereto; and that comprises an agonist of the CSR::T1R, for example, without limitation, calcium chloride, perillartine, NDHC, cyclamate, and cinnamonitrile.
  • Using a kit comprising calcium has the advantage of binding to and activating the chimeric protein only, but not the wild-type receptor or the T1 R2 and T1 R3 part of the chimeric protein.
  • the cells further comprise a G-protein for example for calcium signalling.
  • G-proteins are known and described herein-above; the skilled person is aware how to introduce them to the cells if necessary.
  • a very useful chimeric G-protein is Galpha16-gustducin 44.
  • the agonist is provided in suitable concentrations, for example 1 nM to 10 mM, or 0.1 microM to1 milliM, for example 0.1 microM to 100 microM.
  • Useful concentrations are, for example, for calcium chloride 0.2 to 20 mM, for perillartine 5 to 500 ⁇ M, for cinnamonitrile 10 to 1000 ⁇ M, for cyclamate 0.01 to 5 mM, for Neohesperidin Dihydrochalcone (NDHC) 0.033 to 3.3 mM.
  • Optional kit components may include a suitable medium for culturing the recombinant cells provided, and a solid support to grow the cells on, for example, a cell culture dish or microtiter plate, these optional components will be readily available to the skilled person.
  • the kit may be used as follows:
  • (iii) may be performed according to any one of the assays described-herein above, in combination with any one of the detection methods that report receptor activity described herein-above. This may require specifically chosen or adapted recombinant cells, which are also described herein-above.
  • a suitable assay is, for example, the calcium flux assay to determine activation of CSR::T1 R and its change in response to a test agent.
  • the kit may be used to identify an enhancer as follows:
  • a suitable calcium chloride concentration is, for example, from about 0.2 to 20 mM, or 0.5 to 10 mM, or about 1 mM.
  • a modulator identified by a method described herein-above may easily be confirmed by simple sensory experiments using a panel of flavorists or test persons to taste the identified modulators.
  • the compounds are tasted e.g. in water to confirm sweet taste or together with sweet tastants in comparison to a negative control without modulator to confirm a modultor that enhances the sweet taste.
  • Transcriptional reporter assays and most cell-based assays described herein-above are well suited for screening libraries for agents that modulate CSR::T1R activity.
  • the assays may be designed to screen large chemical libraries by automating the assay steps and providing compounds from any convenient source to the assays, which are typically run in parallel (for example in microtiter formats on microtiter plates in robotic assays).
  • Assays may be run in high throughput screening methods that involve providing a combinatorial chemical or peptide library containing a large number of potential modulators. Such libraries are then screened in one or more assays described herein-above to identify those library agents (particular chemical species or subclasses) that display the activity described herein-above.
  • the modulators thus identified can be directly used or may serve as leads to identify further modulators by making and testing derivatives.
  • Synthetic compound libraries are commercially available from a number of companies including Maybridge Chemical Co. (Trevillet, Cornwall, UK), Comgenex (Princeton, N.J.), Brandon Associates (Merrimack, N.H.), and Microsource (New Milford, Conn.).
  • a combinatorial chemical library is a collection of diverse chemical compounds generated by either chemical synthesis or biological synthesis, by combining a number of chemical "building blocks” such as reagents.
  • a linear combinatorial chemical library such as a polypeptide library is formed by combining a set of chemical building blocks (amino acids) in every possible way for a given compound length (i.e., the number of amino acids in a polypeptide compound). Millions of chemical compounds can be synthesized through such combinatorial mixing of chemical building blocks.
  • a rare chemical library is available from Aldrich (Milwaukee, Wis.).
  • libraries include protein/expression libraries, cDNA libraries from natural sources, including, for example, foods, plants, animals, bacteria, libraries expressing randomly or systematically mutated variants of one or more polypeptides, genomic libraries in viral vectors that are used to express the mRNA content of one cell or tissue.
  • a high throughput assay it is possible to screen up to several thousand different modulators or ligands in a single day.
  • each well of a microtiter plate can be used to run a separate assay against a selected potential modulator, or, if concentration or incubation time effects are to be observed, every 5-10 wells can test a single modulator.
  • a single standard microtiter plate can assay about 100 modulators. If 1536 well plates are used, then a single plate can easily assay from about 100 to about 1500 different compounds. It is possible to assay several different plates per day; assay screens for up to about 6,000-20,000 different compounds is possible.
  • test agents may be any agent including small chemical compounds, chemical polymers, biological polymers, peptides, proteins, sugars, carbohydrates, nucleic acids and lipids.
  • An agent can be a synthetic compound, a mixture of compounds, a natural product or natural sample, for example plant extract, culture supernatant, or tissue sample.
  • methyl chavicol As examples of compounds that may modifiy sweet taste there may be mentioned methyl chavicol, Theasaponin E1, Acesulfame K, Alitame, Aspartame, CH 401, Dulcin, Neotame, sodium Cyclamate, Sucralose, Superaspartame, Cynarin, Glycyphyllin, Rebaudioside C, Abrusoside A, Abrusoside B, Abrusoside C, Abrusoside D, Abrusoside E, Apioglycyrrhizin, Araboglycyrrhizin, Baiyunoside, Brazzein, Bryodulcoside, Carnosifloside V, Carnosifloside VI, D.
  • Identified modulators of sweet tastants may include, for example, modulators of artificial sweeteners that are able to elicit a sweet taste sensation.
  • Consumables include food products, beverages, oral care products, and compositions for admixture to such products, in particular flavour compositions.
  • Flavour compositions may be added to processed foods or beverages during their processing, or they may actually be consumables in their own right, e.g. condiments such as sauces and the like.
  • Sweet tastants are particularly interesting in confectionary and other sweet consumables including desserts, but also in savoury and sweet-sour consumables.
  • consumables examples include confectionary products, cakes, cereal products, baker's products, bread products, gums, chewing gums, sauces (condiments), soups, processed foods, cooked fruits and vegetable products, meat and meat products, egg products, milk and dairy products, cheese products, butter and butter substitute products, milk substitute products, soy products, edible oils and fat products, medicaments, beverages, alcoholic drinks, beers, soft drinks, food extracts, plant extracts, meat extracts, condiments, sweeteners, nutraceuticals, pharmaceutical and non-pharmaceutical gums, tablets, lozenges, drops, emulsions, elixirs, syrups and other preparations for making beverages, instant beverages and effervescent tablets.
  • SEQ ID NO:1 and 19 correspond to the nucleotide/nucleic acid sequence encoding the CSR::T1R2 chimeric proteins (-a/-b)
  • SEQ ID NO: 2 and 20 correspond to the polypeptide/amino acid sequence of the CSR::T1R2 chimeric proteins (-a and -b).
  • SEQ ID NO:3 and 21 correspond to the nucleotide/nucleic acid sequence encoding the CSR::T1R3 chimeric proteins (-a and -b)
  • SEQ ID NO: 4 and 22 correspond to the polypeptide/amino acid sequence of the CSR::T1R3 chimeric proteins (-a and - b).
  • the CSR::T1R2 chimeric protein and the CSR::T1R3 chimeric protein form a functional chimeric sweet receptor.
  • the resulting complex may comprise the two -a variants, the two -b variants, or combinations (CSR::T1 R2-a with CSR::T1R3-b or CSR::T1 R2-b with CSR::T1R3-b), or homologous variants of these that retain their function as herein described.
  • the nucleic acid coding for the novel chimeric protein (SEQ ID NO:1 or 3 for variants-a, and SEQ ID NO: 19 and 21 for variants-b) is followed by the HSV tag at the C-terminus (SEQ ID NO:5).
  • the resulting proteins will accordingly comprise the following amino acids: amino acids of SEQ ID NO:1 followed by SEQ ID NO:5, SEQ ID NO:19 followed by SEQ ID NO:5, SEQ ID NO: 3 followed by SEQ ID NO:5, or SEQ ID NO:21 followed by SEQ ID NO:5.
  • T1 R2 and T1R3 subunits of the T1R2/T1R3 receptor complex are given in SEQ ID NO: 7 + 8 for T1R2, and SEQ ID NO: 9 +10 for T1R3.
  • Fluo-4 is a fluorescent indicator for intracellular calcium and allows to determine changes in the calcium concentration, in particular an increase in response to receptor activation occurring after ligand addition.
  • HEK293 cells stably expressing G ⁇ 16-gustducin 44 were used as host cells and transfected with various constructs as described in example 4.
  • Black, clear-bottom 96-well plates were used for all assays. They were seeded the day before with 8500 transfected cells per well and maintained at 37° C overnight in an a growth medium appropriate for the cells used.
  • a growth medium appropriate for the cells used.
  • Dulbecco's Modified Eagle medium containing high glucose, L-glutamine, pyroxidine hydrochloride, and supplemented with10% fetal bovine serum was used for growth and maintenance of the HEK293 cells.
  • the growth medium was discarded and cells were incubated for 1 hour (at 37° C in the dark) with 50 ⁇ l of a calcium assay solution consisting of 1.5 ⁇ M Fluo-4 AM (Molecular ProbesTM, Invitrogen, US) and 2.5 ⁇ M probenicid (Sigma-Aldrich) dissolved in a reduced calcium C1 buffer solution.
  • Reduced calcium C1 buffer solution contains 130 mM NaCl, 5 mM KCl, 10 mM Hepes, 0.5 mM CaCl2 (reduced from 2 mM) and 10 mM glucose (pH 7.4).
  • the plates were washed 5 times with 100 ⁇ l per well of reduced calcium C1 buffer using an automated plate washer (BioTek) and after washing, the plate was further incubated for 30 minutes at room temperature in the dark to allow for complete de-esterification of the Fluo-4-AM.
  • the buffer solutions were discarded, the plate was washed 5 times with 100 ⁇ l reduced calcium C1 wash buffer and finally the cells were reconstituted in 180 ⁇ l of reduced calcium C1 wash buffer.
  • the plate was placed in a FLIPR (fluorescence imaging plate reader (FLIPR-Tetra, Molecular Devices)), and receptor activation was initiated following addition of 20 ⁇ l of a 10X concentrated ligand stock solution, which were prepared in reduced calcium C1 buffer.
  • FLIPR fluorescence imaging plate reader
  • the CSR::T1 R2-a chimeric cDNA vector construct was generated by joining two DNA fragments generated by PCR via a common restriction enzyme site in both PCR products , namely of a PCR product representing the extracellular amino terminal domain (ATD) of hCaSR (1 - Phe 539 ) to a PCR product representing an "-a" fragment of T1 R2 containing the cysteine-rich domain (CRD), transmembrane (TMD) and C-terminus beginning at Ser 493 (T1 R2-a, SEQ ID No: 1 (nucleic acid) and 2 (protein)).
  • a Sac II site was introduced to the primers that were used to form the two fragments described hereinabove. Using these introduced sites and the appropriate restriction enzyme in buffers and under conditions well known in the art, the fragments were joined by enzymatic ligation.
  • Sac II sites in the formed PCR-products/fragments are located at the C-terminal end of the hCaSR ATD fragment and the N-terminal end of the T1 R2-a fragment, respectively, allowing for ligation of the two PCR-products/fragments of the chimeric DNA. Incorporation of this Sac II site converts Phe 539 in the hCaSR into an arginine residue.
  • PCR using Platinum Taq High Fidelity Polymerase was used to amplify the fragments that comprise the CSR:T1 R2-a chimeric cDNA fragment using the specific primers of SEQ ID 13-16 which are given below. F designates the forward primer, R the reverse primer.
  • the underligned letters designate restriction sites located within the primers for subsequent subcloning of the PCR products.
  • the template for the PCR amplification was a full length cDNA for either the human CaSR (commercially available from Origene Inc., USA), or the human T1 R2, which was isolated from a cDNA library generated from human fungiform papillae taste tissue.
  • PCR reaction parameters were: 94° C for 5 min followed by 35 cycles of 94° C for 45 seconds, 54° C for 15 seconds and 72 ° C for 2 minutes, followed by a final extension cycle of 72° C for 10 minutes.
  • the resulting nucleic acid fragments were separated by gel electrophoresis, purified and subcloned into the pCR-Topo-II vector (Invitrogen) and the resulting clones were verified by DNA sequencing to ensure absence of mutations arising from the PCR amplification. After sequencing, the inserts were subcloned into an expression cassette vector construct based on the pcDNA4-TO vector (purchased from Invitrogen, USA) via 3-piece ligation, allowing for assembly of the CSR::T1 R2-a chimeric cDNA fragment in the vector construct.
  • the C-terminus of the formed vector construct encodes the herpes simplex virus (HSV) glycoprotein D epitope, which can be used for immuncytochemistry studies using a specific antibody that binds to this epitope.
  • HSV herpes simplex virus
  • the resulting CSR:T1 R2-a vector construct with CSR:T1 R2-a cDNA allows for expression of the CSR:T1 R2-a:HSV protein of joined amino acid sequences of Seq ID NO:2 (CSR:T1 R2-a) followed by Seq ID NO:6 (HSV epitope) (in amino terminus to C terminus direction).
  • the CSR::T1 R2-b chimeric cDNA vector construct was generated by joining two DNA fragments generated by PCR via a common restriction enzyme site in both PCR products , namely of a PCR product representing the extracellular amino terminal domain (ATD) and cysteine-rich domain (CRD) of hCaSR (1-IIe 603 ) to a PCR product representing a "-b" fragment of T1 R2 containing the transmembrane (TMD) and C-terminus beginning at Val 557 (T1R2-b, SEQ ID No: 19 (nucleic acid) and 20 (protein)).
  • a BsiW I site was introduced to the primers that were used to form the two fragments described hereinabove. Using these introduced sites and the appropriate restriction enzyme in buffers and under conditions well known in the art, the fragments were joined by enzymatic ligation.
  • BsiW I sites in the formed PCR-products/fragments are located at the C-terminal end of the hCaSR ATD fragment and the N-terminal end of the T1 R2-b fragment, respectively, allowing for ligation of the two PCR-products/fragments of the chimeric DNA. Incorporation of this BsiW I site converts Glu 602 /Ile 603 in the hCaSR into an Arg/Thr residues.
  • PCR using Platinum Taq High Fidelity Polymerase was used to amplify the fragments that comprise the CSR:T1 R2-b chimeric cDNA fragment using the specific primers of SEQ ID 13, 16, 23 and 24 which are given below. F designates the forward primer, R the reverse primer.
  • the underligned letters designate restriction sites located within the primers for subsequent subcloning of the PCR products.
  • the template for the PCR amplification was a full length cDNA for either the human CaSR (commercially available from Origene Inc., USA), or the human T1 R2, which was isolated from a cDNA library generated from human fungiform papillae taste tissue.
  • PCR reaction parameters were: 94° C for 5 min followed by 35 cycles of 94° C for 45 seconds, 54° C for 15 seconds and 72 ° C for 2 minutes, followed by a final extension cycle of 72° C for 10 minutes.
  • the resulting nucleic acid fragments were separated by gel electrophoresis, purified and subcloned into the pCR-Topo-11 vector (Invitrogen) and the resulting clones were verified by DNA sequencing to ensure absence of mutations arising from the PCR amplification.
  • the inserts were subcloned into an expression cassette vector construct based on the pcDNA4-TO vector (purchased from Invitrogen, USA) via 3-piece ligation, allowing for assembly of the CSR::T1 R2-b chimeric cDNA fragment in the vector construct.
  • the C-terminus of the formed vector construct encodes the herpes simplex virus (HSV) glycoprotein D epitope, which can be used for immuncytochemistry studies using a specific antibody that binds to this epitope.
  • HSV herpes simplex virus
  • the resulting CSR:T1 R2-b vector construct with CSR:T1 R2-b cDNA allows for expression of the CSR:T1 R2-b:HSV protein of joined amino acid sequences of Seq ID NO:20 (CSR:T1 R2-b) followed by Seq ID NO:6 (HSV epitope) (in amino terminus to C terminus direction).
  • the CSR::T1 R3-a chimeric cDNA vector construct was generated by joining two DNA fragments generated by PCR via a common restriction enzyme site in both PCR products, namely the joining of a PCR product representing the extracellular amino terminal domain (ATD) of hCaSR (1 - Phe 539 ) to a fragment of T1 R3 containing the cysteine-rich domain (CRD), transmembrane (TMD) and C-terminus beginning at Ser 497 .
  • ATD extracellular amino terminal domain
  • CCD cysteine-rich domain
  • TMD transmembrane
  • Sac II sites in the formed PCR-products/fragments are located at the C-terminal end of the hCaSR-ATD fragment and the N-terminal end of the T1 R3-a fragment, respectively, allowing for ligation of the two fragments. Incorporation of this Sac II site results in a vector construct that comprises a sequence wherein the Phe 539 of the previous hCaSR is converted into an arginine residue. Using the introduced ligation sites and the appropriate restriction enzyme in buffers and under conditions well known in the art, the fragments were joined by enzymatic ligation.
  • PCR using Platinum Taq High Fidelity Polymerase was used to amplify the fragments that comprise the CSR::T1 R3-a chimeric cDNA fragment using the specific primers of Seq ID NO: 17 and Seq ID NO:18 listed below.
  • the amplified PCR-products of T1 R3-a and the amplified PCR products of hCaSR were ligated via the restriction sites indicated in the primer listed below.
  • F designates the forward primer, R the reverse primer.
  • the underligned letters designate restriction sites located within the primers for subsequent ligation and subcloning of the amplified PCR products.
  • the template for the PCR amplification was a full length cDNA for either the hCaSR (purchased from Origene Inc., USA), or the hT1 R3, which was isolated from a cDNA library generated from human fungiform papillae taste tissue.
  • PCR reaction parameters were: 94° C for 5 min followed by 35 cycles of 94° C for 45 seconds, 54° C for 15 seconds and 72 ° C for 2 minutes, followed by a final extension cycle of 72° C for 10 minutes.
  • the resulting nucleic acid fragments (ligation is performed later after the fragments are verified) were separated by gel electrophoresis, purified and subcloned into the pCR-Topo-II vector (Invitrogen, USA). The resulting clones were verified by DNA sequencing to ensure absence of mutations arising from the PCR amplification.
  • the inserts were subcloned into an expression cassette vector construct based on the pcDNA4-TO vector (purchased from Invitrogen, USA) via 3-piece ligation, forming the CSR::T1 R3-a vector construct.
  • the C-terminus of the formed vector construct encodes the herpes simplex virus (HSV) glycoprotein D epitope, which can be used for immuncytochemistry studies using a specific antibody that binds to this epitope.
  • HSV herpes simplex virus
  • the resulting vector construct allows for expression of the CSR::T1 R3-a::HSV protein of joined amino acid sequences of Seq ID NO:4 (CSR::T1R3-a) followed by Seq ID NO:6 (HSV epitope) (in amino terminus to C terminus direction).
  • the CSR::T1 R3-b chimeric cDNA vector construct was generated by joining two DNA fragments generated by PCR via a common restriction enzyme site in both PCR products, namely the joining of a PCR product representing the extracellular amino terminal domain (ATD) and cysteine-rich domain (CRD) of hCaSR (1 - IIe 603 ) to a PCR product representing a fragment of T1 R3 containing the transmembrane (TMD) and C-terminus beginning at Arg 560 ,
  • a BsiW I site was introduced into the primers which were used to make the above-described two fragments.
  • BsiW I sites in the formed PCR-products/fragments are located at the C-terminal end of the hCaSR-ATD fragment and the N-terminal end of the T1 R3-b fragment, respectively, allowing for ligation of the two fragments. Incorporation of this BsiW I site results in a vector construct that comprises a sequence wherein the Phe 539 of the previous hCaSR is converted into an arginine residue. Using the introduced ligation sites and the appropriate restriction enzyme in buffers and under conditions well known in the art, the fragments were joined by enzymatic ligation.
  • PCR using Platinum Taq High Fidelity Polymerase was used to amplify the fragments that comprise the CSR::T1R3-b chimeric cDNA fragment using the specific primers of Seq ID NO: 25 and Seq ID NO:18 listed below.
  • the amplified PCR-products of T1R3-b and the amplified PCR products of hCaSR were ligated via the restriction sites indicated in the primer listed below.
  • F designates the forward primer, R the reverse primer.
  • the underligned letters designate restriction sites located within the primers for subsequent ligation and subcloning of the amplified PCR products.
  • the template for the PCR amplification was a full length cDNA for either the hCaSR (purchased from Origene Inc., USA), or the hT1R3-b, which was isolated from a cDNA library generated from human fungiform papillae taste tissue.
  • PCR reaction parameters were: 94° C for 5 min followed by 35 cycles of 94° C for 45 seconds, 54° C for 15 seconds and 72 ° C for 2 minutes, followed by a final extension cycle of 72° C for 10 minutes.
  • the resulting nucleic acid fragments (ligation is performed later after the fragments are verified) were separated by gel electrophoresis, purified and subcloned into the pCR-Topo-II vector (Invitrogen, USA). The resulting clones were verified by DNA sequencing to ensure absence of mutations arising from the PCR amplification.
  • the inserts were subcloned into an expression cassette vector construct based on the pcDNA4-TO vector (purchased from Invitrogen, USA) via 3-piece ligation, forming the CSR::T1R3-b vector construct.
  • the C-terminus of the formed vector construct encodes the herpes simplex virus (HSV) glycoprotein D epitope, which can be used for immuncytochemistry studies using a specific antibody that binds to this epitope.
  • HSV herpes simplex virus
  • the resulting vector construct allows for expression of the CSR::T1R3-b::HSV protein of joined amino acid sequences of Seq ID NO:22 (CSR::T1R3-b) followed by Seq ID NO:6 (HSV epitope) (in amino terminus to C terminus direction).
  • T1 R2 T1 R3 vector constructs (wildtype receptors for comparison)
  • T1 R2 and T1 R3 vector construct To form the T1 R2 and T1 R3 vector construct, cDNA fragments containing the entire protein coding sequences for human T1 R2 and T1 R3 were isolated from a human fungiform cDNA library, fully sequenced and then subcloned into pCDNA3.1 (Invitrogen).
  • Transfected vector constructs used were those described in examples 2a and 3a, or 2b and 3b, and 4, formed as described above.
  • hCaSR a commercially available pCMV-based vector construct which is based on the full length cDNA was used (TRUECLONE collection, Origene Inc., USA).
  • HEK293T cells that stably express G ⁇ 16-gustducin 44 (formed as described in WO 2004/055048 ) were transfected with the CSR::T1R2 and CSR::T1R3 vector constructs, or with T1 R2 and T1 R3, or hCaSR as follows:
  • the HEK293T/ G ⁇ ,16-gustducin 44 cells were plated in 96-well black, clear-bottom plates at a density of 8500 cells per well and grown overnight in selective growth media.
  • the media was changed to an antibiotic-free and serum-free growth medium and the cells were transfected using 75 ng each of CSR:T1 R2 (-a or -b) and CSR:T1 R3 (total 150 ng) (-a or -b), T1 R2 and T1 R3 (total 150 ng), or 75 ng hCaSR vector construct DNA and 0.3 ⁇ l of Lipofectamine 2000 (Invitrogen).
  • the hCaSR vector is used as positive control for a GPCR that is sensitive to calcium, as it is sensitive to calcium and the calcium binding site lies in the VFT of this receptor, which is where the VFT for the chimera is derived from.
  • CSR:T1 R2/CSR:T1 R3 For transfection of either the CSR:T1 R2/CSR:T1 R3 (CSR:T1 R2-a/CSR:T1 R3-a or CSR:T1 R2-b/CSR:T1 R3-b) or T1R2/T1R3 heterodimers, 75 ng of each vector construct was combined for a total of 150 ng per pair and used together with 0.3 ⁇ l of Lipofectamine 2000. 75 ng of hCaSR vector DNA was used for this calcium-sensing monomeric GPCR.
  • the cells transiently transfected with one of the above-described vector constructs were identified using a fluorescence imaging plate reader (FLIPR-Tetra, Molecular Devices) as described in example 1.
  • a stable cell line was generated in which CSR::T1R3-a was constitutively over-expressed in the presence of a tetracycline-regulated CSR::T1R2-a to avoid possible cytotoxic effects of constitutive over-expression of both proteins.
  • DNA encoding one subunit of the heterodimer (CSR::T1R2-a) was placed in a tetracycline-regulated vector to allow regulation of its expression level so that viability and functionality of the stable clonal lines can be optimised.
  • Human cell lines that stably express the chimeric human CSR::T1R2-a/CSR::T1R3-a heterodimer were generated sequentially by first transfecting a linearized pcDNA4-TO vector (Invitrogen) containing the human CSR::T1R2-a into a G ⁇ 16gust44 expressing cell line, which was prepared as described in WO 2004/055048 .
  • the G ⁇ 16gust44 expressing cell line shows enhanced coupling to taste receptors, is tetracycline inducible, stably expresses the G ⁇ 16gust44 promiscuous G-protein, and is based on the HEK-293-T-Rex cell line (commercially available from Invitrogen, USA).
  • a clonal cell line expressing CSR:T1R2-a was identified and transfected with a linearized pcDNA3.1-Hygro vector (Invitrogen) containing human CSR::T1R3-a cDNA to obtain a double stable clonal cell line that expresses both CSR::T1R2-a and CSR::T1R3-a.
  • the lipofectamine/DNA mixture was incubated on the cells for 3-4 hours and then replaced with an antibiotic-free, serum-containing growth medium. After 24 hours the cells were re-plated in selective medium containing DMEM supplemented with 10% FBS, 0.005 mg/ml blasticidin, 0.36 mg/ml G418, 0.2 mg/ml zeocin, and 0.2 mg/ml hygromycin at 37°C.
  • Resistant colonies were expanded, and identified as containing the CSR::T1R2-a/CSR::T1R3-a heterodimer on the basis of a response to both perillartine (contributed by binding/activation of CSR::T1R2-a) and to sodium cyclamate (contributed by binding/activation of CSR:T1R3-a), which was determined via automated fluorimetric imaging on the FLIPR-Tetra instrumentation (Molecular Devices) using the methods described in example 1. All potential clones were evaluated for a functional response to sweet tastants following induction with 10 ⁇ g/ml tetracycline (to induce over-expression of CSR::T1R2-a).
  • clones were also tested in the absence of tetracycline induction to identify any clones that basally express T1R2-a at a low level, but have sufficient expression of the CSR::T1R2-a receptor to allow for assembly with CSR::T1R3-a resulting in a functional heterodimer complex (the tetracycline-regulated systems such as the T-Rex HEK-293 (Invitrogen) are known to have a low-level basal expression of transgenes due to the inherent leakiness of the system).
  • the tetracycline-regulated systems such as the T-Rex HEK-293 (Invitrogen) are known to have a low-level basal expression of transgenes due to the inherent leakiness of the system).
  • Stable clones expressing an inducible functional CSR::T1R2-a/CSR::T1R3-a heterodimer were identified on the basis of a response to both_50 microM perillartine and to 5 mM (milliM) sodium cyclamate.
  • One clonal cell line exhibiting the greatest tetracycline-inducible response to multiple sweet tastants was propagated and used for subsequent comparisons. The results for tests with various ligands/sweet tastants are indicated in the table below.
  • the ⁇ F/F value obtained corresponds to the calcium increase of the cell in response to a direct or indirect interaction with the transfected receptor ("signal") (the mean (AVG) and standard deviation (S.D.) of three replicate experiments is given).
  • p-ETBZ p-ethoxybenzaldehyde NDHC Sodium Cyclamate (Cyclamate is a sweet tastant. The additional presence of sodium ions improves water solubility, but does not further contribute to sweet taste)
  • the intracellular calcium response following stimulation with various ligands was determined in HEK293T cells stably expressing G ⁇ 16-gustducin 44 and transfected with CSR::T1R2-a/CSR::T1 R3-a chimeric heterodimer.
  • the results were compared to results obtained in cells transfected with the both the T1 R2 vector construct and the T1 R3 vector construct which is described in example 4 (to form the wildtype T1 R2/T1 R3 sweet heterodimer) or the hCaSR vector construct described in example 5 (to form monomeric hCaSR).
  • Mock transfected HEK293T/ G ⁇ 16-gustducin 44 cells transfected without construct that do not express a sweet receptor were used as a negative control to determine signals corresponding merely to the background.
  • the transfected cells are exposed to the sweeteners as indicated and to a positive control (calcium) for the proteins containing Calcium-sensing domains, and to a negative control (C1 buffer).
  • the negative control/mock transfection shows the signal level corresponding to background signals.
  • the hCaSR responded only to calcium chloride and could not be activated by any of the sweet tastants tested.
  • CSR::T1R2-a/CSR::T1R3-a is activated by calcium, perillartine, cyclamate, cinnamonitrile, and neohesperidin dihydrochalcone (NDHC) but not by sucralose or aspartame.
  • the intracellular calcium response following stimulation with various ligands was determined in HEK293T cells stably expressing G ⁇ 16-gustducin 44 and transfected with CSR::T1R2-b/CSR::T1R3-b chimeric heterodimer.
  • the results were compared to results obtained in cells transfected with the both the T1 R2 vector construct and the T1 R3 vector construct which is described in example 4 (to form the T1 R2/T1 R3 sweet heterodimer) or the hCaSR vector construct described in example 5 (to form monomeric hCaSR).
  • the calcium mobilization signals obtained are increase in peak fluorescence (F) normalized to the baseline fluorescence (F 0 ).
  • the ⁇ F/F value obtained corresponds to the calcium increase of the cell in response to a direct or indirect interaction with the transfected receptor ("signal").
  • Mock transfected HEK293T/ G ⁇ 16-gustducin 44 cells transfected without construct that do not express a sweet receptor were used as a negative control to determine signals corresponding merely to the background.
  • the transfected cells are exposed to the sweeteners as indicated and to a positive control (calcium) for the proteins containing Calcium-sensing domains, and to a negative control (C1 buffer).
  • the AVG column gives the mean ⁇ F/F
  • the STD column gives the standard deviation.
  • Table 2b Results CSR::T1R2-b/ CSR::T1R3-b CSR::R2-b alone T1R2/T1R3 Neg.
  • the negative control/mock transfection shows the signal level corresponding to background signals.
  • sucralose For sucralose, a signal is detected in the cells transfected with the T1 R2/1 R3 heterodimer (wildtype) only. Sucralose is believed to bind in the VFT of T1 R2, which is absent in the CSR::T1R chimerae, which explains the lack of signal in the CSR::T1 R2-b/CSR::T1 R3-b heterodimer.
  • the hCaSR responded only to calcium chloride and could not be activated by any of the sweet tastants tested.
  • receptors, nucleic acids, polypeptides, methods and kit have been described above in connection with certain illustrative embodiments, it is to be understood that other similar embodiments may be used or modifications and additions may be made to the described embodiments for performing the same function(s). Further, all embodiments disclosed are not necessarily in the alternative, as various embodiments may be combined to provide the desired characteristics. Therefore, the receptors, nucleic acids, polypeptides, methods and kit should not be limited to any single embodiment, but rather construed in breadth and scope in accordance with the recitation of the attached claims.

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Engineering & Computer Science (AREA)
  • Immunology (AREA)
  • Molecular Biology (AREA)
  • Biomedical Technology (AREA)
  • Hematology (AREA)
  • Urology & Nephrology (AREA)
  • Biochemistry (AREA)
  • Medicinal Chemistry (AREA)
  • General Health & Medical Sciences (AREA)
  • Cell Biology (AREA)
  • Organic Chemistry (AREA)
  • Toxicology (AREA)
  • Microbiology (AREA)
  • Analytical Chemistry (AREA)
  • Physics & Mathematics (AREA)
  • General Physics & Mathematics (AREA)
  • Pathology (AREA)
  • Food Science & Technology (AREA)
  • Biotechnology (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Genetics & Genomics (AREA)
  • Biophysics (AREA)
  • Gastroenterology & Hepatology (AREA)
  • Zoology (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Tropical Medicine & Parasitology (AREA)
  • Micro-Organisms Or Cultivation Processes Thereof (AREA)
  • Peptides Or Proteins (AREA)
  • Measuring Or Testing Involving Enzymes Or Micro-Organisms (AREA)
  • Preparation Of Compounds By Using Micro-Organisms (AREA)

Claims (25)

  1. Protéine chimère CSR::T1 R étant capable de se lier à au moins un édulcorant ou un exhausteur de sucrosité, comprenant une ou plusieurs CSR::T1 R choisie(s) dans le groupe constitué par
    une CSR::T1 R2 de SEQ ID n° : 2 ou SEQ ID n° : 20 ou bien un polypeptide homologue à la SEQ ID n° : 2 ou SEQ ID n° : 20 avec une identité de séquence d'au moins 90%,
    une CSR::T1 R3 de SEQ ID n° : 4 ou SEQ ID n° : 22 ou bien un polypeptide homologue à la SEQ ID n° : 4 ou SEQ ID n° : 22 avec une identité de séquence d'au moins 90%.
  2. Protéine hétérodimérique comprenant une protéine chimère CSR::T1R, selon la revendication 1, choisie dans le groupe constitué par :
    une protéine chimère hétérodimérique CSR::T1R2/CSR::T1R3, une protéine chimère hétérodimérique CSR::T1R2/T1R3 et une protéine chimère hétérodimérique T1R2/CSR::T1R3,
    dans laquelle la sous-unité T1R2 de l'hétérodimère comprend un polypeptide qui est substantiellement homologue à la SEQ ID n° : 8, avec une identité de séquence d'au moins 90% ;
    et dans laquelle la sous-unité T1R3 de l'hétérodimère comprend un polypeptide qui est substantiellement homologue à la SEQ ID n° : 10, avec une identité de séquence d'au moins 90%.
  3. Protéine chimère CSR::T1R comprenant deux sous-unités polypeptidiques, selon la revendication 2, qui comprennent une protéine chimère hétérodimérique CSR::T1R2/CSR::T1R3, y compris une protéine hétérodimérique CSR::T1R2-a/CSR::T1R3-a, une protéine hétérodimérique CSR::T1R2-b/CSR::T1R3-b, une protéine hétérodimérique CSR::T1R2-a/CSR::T1R3-b, une protéine hétérodimérique CSR::T1R2-b/CSR::T1R3-a ou une protéine hétérodimérique étant substantiellement homologue à celles-ci mais sans être limitée à celles-ci, dans laquelle CSR::T1R2-a correspond à la SEQ ID n° : 2, CSR::T1R2-b correspond à la SEQ ID n° : 20, CSR::T1R3-a correspond à la SEQ ID n° : 4 et CSR::T1R3-b correspond à la SEQ ID n° : 22.
  4. Acide nucléique codant pour une protéine chimère CSR::T1R étant capable de se lier à au moins un édulcorant ou un exhausteur de sucrosité, comprenant un ou plusieurs élément(s) parmi
    un acide nucléique étant substantiellement homologue à une séquence nucléotidique choisie dans le groupe constitué par la SEQ ID n° : 1 (CSR::T1R2-a), la SEQ ID n° : 19 (CSR::T1R2-b), la SEQ ID n° : 3 (CSR::T1R3-a) et la SEQ ID n° : 21 (CSR::T1R3-b), tel que déterminé par identité de séquence,
    un acide nucléique étant substantiellement homologue à une séquence nucléotidique choisie dans le groupe constitué par la SEQ ID n° : 1 (CSR::T1R2-a), la SEQ ID n° : 19 (CSR::T1R2-b), la SEQ ID n° : 3 (CSR::T1R3-a) et la SEQ ID n° : 21 (CSR::T1R3-b), tel que déterminé par hybridation,
    un acide nucléique étant substantiellement homologue à une séquence nucléotidique qui code pour la protéine chimère CSR::T1R telle que définie dans la revendication 1,
    dans lequel l'acide nucléique substantiellement homologue, tel que déterminé par identité de séquence, a une identité de séquence d'au moins 90% ;
    dans lequel l'acide nucléique substantiellement homologue, tel que déterminé par hybridation, s'hybride dans des conditions stringentes d'hybridation à une température de 42°C dans une solution constituée par du formamide à 50%, du 5x SSC et du SDS à 1%, ainsi qu'un lavage à 65°C dans une solution constituée par du 0,2x SSC et du SDS à 0,1% ;
    dans lequel l'acide nucléique comprend, en option, la SEQ ID n° : 6 (étiquette du HSV), au niveau de son extrémité ou à la fin de celle-ci, pour former l'extrémité C-terminale dans la protéine correspondante.
  5. Vecteur d'expression comprenant l'acide nucléique tel que défini dans la revendication 4.
  6. Cellule hôte transfectée avec un vecteur d'expression tel que défini dans la revendication 5.
  7. Cellule hôte, selon la revendication 6, exprimant, de manière stable, une protéine chimère CSR::T1R, telle que définie dans la revendication 1, et une protéine G, facultativement une protéine G qui est substantiellement homologue à la Gaq-Gustducine.
  8. Cellule hôte, selon la revendication 6, exprimant, de manière transitoire, une protéine chimère CSR::T1R, telle que définie dans la revendication 1, et une protéine G, facultativement une protéine G qui est substantiellement homologue à la Gaq-Gustducine.
  9. Procédé de production d'une protéine chimère CSR::T1R, telle que définie dans les revendications 1, 2 ou 3, comprenant les étapes consistant à cultiver des cellules hôtes, comprenant un vecteur d'expression qui code pour la protéine chimère CSR::T1R, dans des conditions suffisantes pour l'expression, en formant de ce fait la protéine chimère CSR::T1R, et facultativement en la récupérant à partir des cellules.
  10. Procédé d'identification d'un agent qui module la signalisation du goût sucré dans les cellules du goût, le procédé comprenant les étapes consistant à :
    (i) mettre en contact les cellules exprimant une protéine chimère CSR::T1R, qui répond à un stimulus choisi parmi un stimulus du goût sucré et un stimulus au calcium, avec un agent en fournissant de ce fait une réponse fonctionnelle, facultativement en présence d'un autre agent ; et
    (ii) déterminer si au moins un agent affecte la réponse fonctionnelle de ladite protéine chimère CSR::T1R dans lesdites cellules, par au moins une réponse fonctionnelle dans lesdites cellules ;
    dans lequel ladite protéine chimère CSR::T1R est telle que définie dans les revendications 1, 2 ou 3.
  11. Procédé selon la revendication 10, dans lequel les cellules expriment également une protéine G.
  12. Procédé selon la revendication 11, dans lequel la protéine G est une protéine G chimère étant substantiellement homologue à la Gaq-Gustducine.
  13. Procédé selon la revendication 12, dans lequel la protéine G est la protéine G chimère G alpha 16-gustducine 44.
  14. Procédé selon la revendication 10, dans lequel l'étape (ii) est exécutée en mesurant un changement dans les messagers intracellulaires ou étant causé par ceux-ci.
  15. Procédé selon la revendication 11, dans lequel la réponse fonctionnelle est déterminée en mesurant un changement dans un messager intracellulaire choisi parmi l'IP3 et le calcium2+.
  16. Procédé selon la revendication 10, dans lequel lesdites cellules sont choisies dans le groupe constitué par des cellules bactériennes, cellules eucaryotes, cellules de levure, cellules d'insecte, cellules de mammifère, cellules d'amphibien et cellules de ver.
  17. Procédé selon la revendication 16, dans lequel la cellule est une cellule de mammifère.
  18. Procédé selon la revendication 17, dans lequel la cellule est une cellule de mammifère choisie dans le groupe constitué par les cellules CHO, COS, HeLa et HEK-293.
  19. Procédé selon la revendication 10, dans lequel l'étape (i) comprend en outre la mise en contact de la protéine chimère CSR::T1R avec un agent de test en présence de calcium.
  20. Procédé selon la revendication 19, dans lequel le calcium est fourni sous forme de chlorure de calcium.
  21. Trousse comprenant :
    (i) des cellules recombinantes qui expriment une protéine chimère CSR::T1R, telle que définie dans les revendications 1, 2 ou 3, et
    (ii) un agoniste de la protéine chimère CSR::T1R, destinés à une utilisation combinée afin d'identifier des agents de test en tant que modulateurs de la protéine chimère CSR::T1R.
  22. Procédé d'utilisation de la trousse, selon la revendication 21, comprenant les étapes consistant à :
    (i) cultiver des cellules recombinantes qui expriment une protéine chimère CSR::T1R,
    (ii) ajouter des agents de test, en présence de l'agoniste à une concentration appropriée, et (iii)déterminer un changement dans une réponse fonctionnelle des cellules, par comparaison de la réponse en présence et en l'absence de l'agent de test, et l'agent de test est, de ce fait, identifié en tant que modulateur de la protéine chimère CSR::T1R telle que définie dans les revendications 1, 2 ou 3.
  23. Procédé d'identification d'un agent qui module la protéine chimère CSR::T1R, telle que définie dans les revendications 1, 2 ou 3, le procédé comprenant les étapes consistant à :
    (i) mesurer un paramètre qui change en réponse à la liaison d'un ligand à la protéine chimère CSR::T1R, et
    (ii) déterminer un changement du paramètre en réponse à un agent de test, facultativement en présence d'un ligand, en comparaison avec un témoin négatif et en identifiant, de ce fait, un modulateur ou un ligand.
  24. Procédé selon la revendication 23, dans lequel le ligand est choisi dans le groupe constitué par le calcium, des ions calcium et le chlorure de calcium.
  25. Procédé selon l'une quelconque des revendications 23 et 24, dans lequel l'étape (i) est exécutée par un procédé choisi dans le groupe constitué par la spectroscopie par fluorescence, la spectroscopie par RMN, une mesure d'un ou plusieurs élément(s) parmi l'absorbance, l'indice de réfraction, des procédés hydrodynamiques, la chromatographie, une mesure de la solubilité, des procédés biochimiques, dans lequel les procédés mesurent les propriétés de la protéine chimère CSR::T1R dans un environnement approprié, choisi dans le groupe constitué par une solution, une membrane bicouche, fixée à une phase solide, dans une monocouche lipidique, liée à une membrane et dans des vésicules.
EP07720194A 2006-06-19 2007-06-15 Acide nucléique, polypeptide et son utilisation Active EP2035827B1 (fr)

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
US81486606P 2006-06-19 2006-06-19
US88140207P 2007-01-19 2007-01-19
PCT/CH2007/000297 WO2007147275A1 (fr) 2006-06-19 2007-06-15 Acide nucléique, polypeptide et son utilisation

Publications (2)

Publication Number Publication Date
EP2035827A1 EP2035827A1 (fr) 2009-03-18
EP2035827B1 true EP2035827B1 (fr) 2012-10-17

Family

ID=38608811

Family Applications (1)

Application Number Title Priority Date Filing Date
EP07720194A Active EP2035827B1 (fr) 2006-06-19 2007-06-15 Acide nucléique, polypeptide et son utilisation

Country Status (5)

Country Link
US (1) US7964376B2 (fr)
EP (1) EP2035827B1 (fr)
JP (1) JP5225986B2 (fr)
KR (1) KR101440639B1 (fr)
WO (1) WO2007147275A1 (fr)

Families Citing this family (17)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US8039233B2 (en) 2007-01-18 2011-10-18 Givaudan S.A. Nucleic acid sequences and their use in methods to identify umami modulatiors
BR112012030836B8 (pt) 2010-06-02 2024-02-06 Evolva Nutrition Inc Hospedeiro recombinante que compreende genes recombinantes para produção de esteviol ou glicosídeo de esteviol, método para produzir esteviol, glicosídeo de esteviol ou composição de glicosídeo de esteviol e método para sintetizar esteviol ou glicosídeo de esteviol
BR122021015509B1 (pt) 2011-08-08 2022-03-29 Evolva Sa Método para produzir um glicosídeo de esteviol alvo
WO2013072332A1 (fr) 2011-11-14 2013-05-23 Givaudan Sa Procédés d'utilisation d'antagonistes de récepteurs du goût amer
CA2899276C (fr) 2013-02-06 2022-11-01 Evolva Sa Procedes pour la production amelioree de rebaudioside d et de rebaudioside m
SG10201705993YA (en) 2013-02-11 2017-08-30 Evolva Sa Efficient production of steviol glycosides in recombinant hosts
AU2014262540B2 (en) 2013-05-10 2016-10-27 Pepsico, Inc. Taste receptor internalization assay
JP6367315B2 (ja) * 2013-05-10 2018-08-01 ペプシコ, インコーポレイテッドPepsiCo Inc. 潜在的味覚調節物質を特定するためのモデルとしての細胞
BR112017002783A2 (pt) 2014-08-11 2017-12-19 Evolva Sa produção de glicosídeos de esteviol em hospedeiros recombinantes
MX2017003130A (es) 2014-09-09 2017-10-24 Evolva Sa Produccion de glucosidos de esteviol en hospederos recombinantes.
CA2973674A1 (fr) 2015-01-30 2016-08-04 Evolva Sa Production de glycosides de steviol dans des hotes de recombinaison
CN115851470A (zh) 2015-03-16 2023-03-28 帝斯曼知识产权资产管理有限公司 Udp-糖基转移酶
AU2016307066A1 (en) 2015-08-07 2018-02-08 Evolva Sa Production of steviol glycosides in recombinant hosts
US10982249B2 (en) 2016-04-13 2021-04-20 Evolva Sa Production of steviol glycosides in recombinant hosts
CN109312378A (zh) 2016-05-16 2019-02-05 埃沃尔瓦公司 在重组宿主中产生甜菊醇糖苷
US11396669B2 (en) 2016-11-07 2022-07-26 Evolva Sa Production of steviol glycosides in recombinant hosts
KR102624889B1 (ko) * 2021-11-30 2024-01-15 서울대학교산학협력단 트립토판 형광 분석을 이용한 인간 단맛 수용체의 리간드 결합 도메인의 맛 물질에 대한 리간드 효능 분석 방법

Family Cites Families (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP1585954B1 (fr) * 2001-07-03 2014-03-26 The Regents of The University of California Recepteurs de gout heterodimeriques de gout sucre et d'acide amine, chez les mammiferes
WO2005041684A2 (fr) * 2003-08-06 2005-05-12 Senomyx Inc. Saveurs, modificateurs de saveur, agents gustatifs, rehausseurs de gout, agents gustatifs et/ou rehausseurs umami ou sucres, et utilisation correspondante

Also Published As

Publication number Publication date
KR101440639B1 (ko) 2014-09-22
US20090286262A1 (en) 2009-11-19
EP2035827A1 (fr) 2009-03-18
KR20090029218A (ko) 2009-03-20
JP2009540804A (ja) 2009-11-26
JP5225986B2 (ja) 2013-07-03
US7964376B2 (en) 2011-06-21
WO2007147275A1 (fr) 2007-12-27

Similar Documents

Publication Publication Date Title
JP7292347B2 (ja) 甘味増強に関する方法
EP2035827B1 (fr) Acide nucléique, polypeptide et son utilisation
US8546085B2 (en) Methods of identifying modulators of the bitter taste receptor TAS2R44
US8597896B2 (en) Methods to identify modulators using TAS2R bitter taste receptor
US8124360B2 (en) Use of a T1R2 nucleic acid sequence to identify tastants
US20110097741A1 (en) Partial t1r2 nucleic acid sequence, receptor protein and its use in screening assays
EP2329271B1 (fr) Procédés d'identification de modulateurs
US8039233B2 (en) Nucleic acid sequences and their use in methods to identify umami modulatiors

Legal Events

Date Code Title Description
PUAI Public reference made under article 153(3) epc to a published international application that has entered the european phase

Free format text: ORIGINAL CODE: 0009012

17P Request for examination filed

Effective date: 20081216

AK Designated contracting states

Kind code of ref document: A1

Designated state(s): AT BE BG CH CY CZ DE DK EE ES FI FR GB GR HU IE IS IT LI LT LU LV MC MT NL PL PT RO SE SI SK TR

AX Request for extension of the european patent

Extension state: AL BA HR MK RS

17Q First examination report despatched

Effective date: 20100311

REG Reference to a national code

Ref country code: DE

Ref legal event code: R079

Ref document number: 602007026133

Country of ref document: DE

Free format text: PREVIOUS MAIN CLASS: G01N0033500000

Ipc: C07K0014705000

RIC1 Information provided on ipc code assigned before grant

Ipc: C07K 14/435 20060101ALI20120216BHEP

Ipc: C07K 14/705 20060101AFI20120216BHEP

Ipc: G01N 33/50 20060101ALN20120216BHEP

RIC1 Information provided on ipc code assigned before grant

Ipc: C07K 14/705 20060101AFI20120220BHEP

Ipc: C07K 14/435 20060101ALI20120220BHEP

Ipc: G01N 33/50 20060101ALN20120220BHEP

RIC1 Information provided on ipc code assigned before grant

Ipc: C07K 14/705 20060101AFI20120229BHEP

Ipc: G01N 33/50 20060101ALN20120229BHEP

Ipc: C07K 14/435 20060101ALI20120229BHEP

GRAP Despatch of communication of intention to grant a patent

Free format text: ORIGINAL CODE: EPIDOSNIGR1

DAX Request for extension of the european patent (deleted)
GRAS Grant fee paid

Free format text: ORIGINAL CODE: EPIDOSNIGR3

GRAA (expected) grant

Free format text: ORIGINAL CODE: 0009210

AK Designated contracting states

Kind code of ref document: B1

Designated state(s): AT BE BG CH CY CZ DE DK EE ES FI FR GB GR HU IE IS IT LI LT LU LV MC MT NL PL PT RO SE SI SK TR

REG Reference to a national code

Ref country code: GB

Ref legal event code: FG4D

REG Reference to a national code

Ref country code: CH

Ref legal event code: EP

REG Reference to a national code

Ref country code: IE

Ref legal event code: FG4D

REG Reference to a national code

Ref country code: AT

Ref legal event code: REF

Ref document number: 579884

Country of ref document: AT

Kind code of ref document: T

Effective date: 20121115

Ref country code: CH

Ref legal event code: NV

Representative=s name: GIVAUDAN SCHWEIZ AG GLOBAL PATENTS, CH

REG Reference to a national code

Ref country code: DE

Ref legal event code: R096

Ref document number: 602007026133

Country of ref document: DE

Effective date: 20121213

REG Reference to a national code

Ref country code: NL

Ref legal event code: T3

REG Reference to a national code

Ref country code: AT

Ref legal event code: MK05

Ref document number: 579884

Country of ref document: AT

Kind code of ref document: T

Effective date: 20121017

REG Reference to a national code

Ref country code: LT

Ref legal event code: MG4D

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: LT

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20121017

Ref country code: IS

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20130217

Ref country code: FI

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20121017

Ref country code: SE

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20121017

Ref country code: ES

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20130128

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: PT

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20130218

Ref country code: PL

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20121017

Ref country code: LV

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20121017

Ref country code: CY

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20121017

Ref country code: BE

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20121017

Ref country code: GR

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20130118

Ref country code: SI

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20121017

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: AT

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20121017

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: CZ

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20121017

Ref country code: BG

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20130117

Ref country code: SK

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20121017

Ref country code: EE

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20121017

Ref country code: DK

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20121017

PLBE No opposition filed within time limit

Free format text: ORIGINAL CODE: 0009261

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: NO OPPOSITION FILED WITHIN TIME LIMIT

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: IT

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20121017

Ref country code: RO

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20121017

26N No opposition filed

Effective date: 20130718

REG Reference to a national code

Ref country code: DE

Ref legal event code: R097

Ref document number: 602007026133

Country of ref document: DE

Effective date: 20130718

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: MC

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20121017

REG Reference to a national code

Ref country code: IE

Ref legal event code: MM4A

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: IE

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20130615

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: MT

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20121017

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: TR

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20121017

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: LU

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20130615

Ref country code: HU

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT; INVALID AB INITIO

Effective date: 20070615

REG Reference to a national code

Ref country code: FR

Ref legal event code: PLFP

Year of fee payment: 10

REG Reference to a national code

Ref country code: FR

Ref legal event code: PLFP

Year of fee payment: 11

REG Reference to a national code

Ref country code: FR

Ref legal event code: PLFP

Year of fee payment: 12

REG Reference to a national code

Ref country code: CH

Ref legal event code: NV

Representative=s name: GIVAUDAN INTERNATIONAL SA GLOBAL PATENTS, CH

P01 Opt-out of the competence of the unified patent court (upc) registered

Effective date: 20230421

PGFP Annual fee paid to national office [announced via postgrant information from national office to epo]

Ref country code: NL

Payment date: 20230620

Year of fee payment: 17

Ref country code: FR

Payment date: 20230627

Year of fee payment: 17

PGFP Annual fee paid to national office [announced via postgrant information from national office to epo]

Ref country code: CH

Payment date: 20230702

Year of fee payment: 17

PGFP Annual fee paid to national office [announced via postgrant information from national office to epo]

Ref country code: GB

Payment date: 20240620

Year of fee payment: 18

PGFP Annual fee paid to national office [announced via postgrant information from national office to epo]

Ref country code: DE

Payment date: 20240619

Year of fee payment: 18